<<

International Journal of Molecular Sciences

Review WNT Signaling in

Anna Gajos-Michniewicz and Malgorzata Czyz *

Department of Molecular Biology of , Medical University of Lodz, 6/8 Mazowiecka Street, 92–215 Lodz, Poland; [email protected] * Correspondence: [email protected]; Tel.: +48-42272570

 Received: 22 May 2020; Accepted: 7 July 2020; Published: 9 July 2020 

Abstract: WNT-signaling controls important cellular processes throughout and adult life, so any deregulation of this signaling can result in a wide range of pathologies, including cancer. WNT-signaling is classified into two categories: β--dependent signaling (canonical pathway) and β-catenin-independent signaling (non-canonical pathway), the latter can be further divided into WNT/planar (PCP) and calcium pathways. WNT ligands are considered as unique directional growth factors that contribute to both and polarity. Origin of cancer can be diverse and therefore tissue-specific differences can be found in WNT-signaling between , including specific contributing to cancer development. This review focuses on the role of the WNT-signaling pathway in melanoma. The current view on the role of WNT-signaling in cancer immunity as well as a short summary of WNT pathway-related drugs under investigation are also provided.

Keywords: WNT; β-catenin; WNT5A; melanoma; immune evasion; crosstalk

1. Introduction

The study of WNT-signaling was initiated in the early 19800s by the discovery of Wingless, a segment polarity [1] and then the mouse proto- Int1 [2]. The term ‘WNT’ comes from a combination of these two names of the same gene [3]. The WNT-signaling is evolutionarily conserved and plays an important role in the embryonic development, adult tissue and [4]. Furthermore, it maintains genetic stability and is important for cell fate and differentiation, cell proliferation, cell motility, and stem cell maintenance [5]. Aberrant functioning of WNT-signaling is associated with a number of diseases, including embryonic malformations, degenerative diseases and cancer [6–9]. WNT-signaling is divided into two pathways: β-catenin-dependent also known as canonical or WNT/β-catenin pathway and β-catenin-independent—also termed as non-canonical—which can be further divided into WNT/planar cell polarity (PCP) and calcium pathway that in some circumstances can antagonize WNT/β-catenin-signaling [10]. The β-catenin-dependent pathway mainly controls cell proliferation, whereas β-catenin-independent signaling regulates cell polarity and migration. This distinction, however, is conventional as these two main pathways form a network with concomitant crosstalk and mutual regulation [11,12]. Better understanding of the mechanisms that govern the highly context-dependent outcome of WNT-signaling in different tumors is important for the development of appropriate treatment strategies. This review is focused on WNT-signaling in melanoma, a tumor derived from melanocytes that arise from cells.

1.1. WNT Ligands in Canonical and Non-Canonical WNT Signaling Pathways The WNT family of secreted includes 19 cysteine-rich (~40 kDa; ~350–400 amino acids with a 20–85% sequence identity) [4,13], in which postranslational modifications comprising and are considered to be essential for their biologic activity [6,14]. Porcupine, resident acyltransferase, is the that is required for the

Int. J. Mol. Sci. 2020, 21, 4852; doi:10.3390/ijms21144852 www.mdpi.com/journal/ijms Int. J. Mol. Sci. 2020, 21, 4852 2 of 31 attachment of palmitoleic acid to WNT ligands [6,8,14]. Then, WNT ligands bind to an evolutionary highly conserved transmembrane Evenness interrupted/Wntless (EVI/WLS) and are shuttled to the plasma membrane via the Golgi apparatus [15]. By -mediated endocytosis, EVI/WLS Int. J. Mol. Sci. 2020, 21, x FOR PEER REVIEW 2 of 32 is recycled in the Golgi apparatus by the complex. There are several routes enabling WNT proteins toendoplasmic exit the reticulum cells: by resident solubilization, acyltransferase, exosome is the enzyme formation that is or required by lipoprotein for the attachment particles of (LPPs), serving aspalmitoleic extracellular acid to transporters WNT ligands [6,8,14]. to achieve Then, long-rangeWNT ligands signalingbind to an evolutionary [4,8,15]. The highly interactions conserved between transmembrane protein Evenness interrupted/Wntless (EVI/WLS) and are shuttled to the plasma WNTs andmembrane their specific via the Golgi receptors apparatus activate [15]. ByWNT clathrin pathways:-mediated endocytosis, canonical EVI/WLS (β-catenin-dependent) is recycled in the Golgi (Figure1) and non-canonicalapparatus by ( βthe-catenin-independent) retromer complex. There are (Figure several2 routes) that enabling cooperate WNT with proteins each to exit other the incells: regulation by of importantsolubilization, cellular processes. exosome formation Generally, or by the lipoprotein subtypeparticles (LPPs), determines serving as the extracellular mode of thetransporters WNT-signaling to achieve long-range signaling [4,8,15]. The interactions between WNTs and their specific receptors activate network.WNT WNT1, pathways: WNT2, canonical WNT3, (β-catenin WNT3A,-dependent) WNT8a, (Figure WNT8b, 1) and non WNT10a-canonical and (β-catenin WNT10b-independent) are activators of the canonical(Figure 2) pathway,that cooperate whereas with eachWNT4, other in regulation WNT5A, of important WNT5B, cellular WNT6, processes. WNT7a, Generally, WNT7b the ligand and WNT11 are commonsubtype activators determines of the non-canonical mode of the WNT WNT-signaling-signaling network.[ 16WNT1,,17]. WNT2, WNTs WNT3, are classifiedWNT3A, WNT8a, as directional growth factorsWNT8b, with WNT10a unique and WNT10b properties are activators since of they the canonical influence pathway, proliferation whereas WNT4, and WNT5A, polarity, WNT5B, and both may WNT6, WNT7a, WNT7b and WNT11 are common activators of non-canonical WNT-signaling [16,17]. occur atWNTs the same are classified time andas directional in the growth same factors cells [with18]. unique Moreover, properties WNTs since they can influence act in proliferation an autocrine and paracrineand manner polarity, [6 and,19 ,both20]. may occur at the same time and in the same cells [18]. Moreover, WNTs can act in an autocrine and paracrine manner [6,19,20].

Figure 1. Simplified scheme of canonical WNT -signaling pathway. (A) In the absence of WNT ligands (WNT Figure 1.OFFSimplified state), β-catenin scheme is phosphorylated of canonical by a destruction WNT -signaling complex consisting pathway. of AXIN, (A APC,) In GSK3β the absence and CK1α of WNT ligands (WNTto be further OFF ubiquitina state),tedβ for-catenin proteasomal is phosphorylated degradation. In the absence by a of destruction R-spondins, E3 complex ligases consisting of AXIN, APC,RNF43/ZNRF3 GSK3β targetand FZD CK1 forα lysosomalto be furtherdegradation; ubiquitinated (B) binding of WNT for ligands proteasomal to FZD receptors degradation. and LRP In the co-receptors activates WNT-signaling (WNT ON state). AXIN is associated with LRP5/6, whereas DVL is absence of R-spondins, E3 ubiquitin ligases RNF43/ZNRF3 target FZD for lysosomal degradation; recruited to FZD, which results in dissociation of the destructive complex. β-catenin is accumulated and (B) bindingstabilized of WNT in the ligands cytosol, and to FZDthen unphosphorylated receptors and β LRP-catenin co-receptors is translocated activates to the nucleus WNT-signaling to activate the (WNT ON state).expression AXIN of isWNT associated target . with APC— LRP5adenomatosis/6, whereas polyposis DVL coli; isAXIN recruited—axis inhibition to FZD, protein; which BCL— results in dissociationB-cell of CLL/lymphoma the destructive protein; complex. BRG-1β—-cateninbrahma-related is accumulated gene-1; CBP— and(CREB) stabilized-binding in protein; the cytosol, CK1α— and then 1α; CK1γ—casein kinase 1γ; CK1ε—casein kinase 1ε; DKK1—-1; DVL—disheveled; β unphosphorylatedFZD— -cateninreceptor; GSK3β is translocated—glycogen tosynthase the nucleus kinase 3β; to LEF activate—lymphoid the expression -binding of WNT factor target 1; genes. APC—adenomatosisLGR—leucine-rich polyposis repeat-containing coli; AXIN—axis G-protein coupled inhibition ; protein; LRP— BCL—B-celllow-density lipoprotein CLL/lymphoma receptor protein; BRG-1—brahma-related gene-1; CBP—(CREB)-binding protein; CK1α—casein kinase 1α; CK1γ—casein

kinase 1γ; CK1ε—casein kinase 1ε; DKK1—Dickkopf-1; DVL—disheveled; FZD—frizzled receptor; GSK3β—glycogen synthase kinase 3β; LEF—lymphoid enhancer-binding factor 1; LGR—leucine-rich repeat-containing G-protein coupled receptor; LRP—low-density lipoprotein receptor related protein; MAK— associated kinase; PAR1—protease-activated receptor 1; PKC— C; PYGO—pygopus; RNF43—ring finger protein 43; sFRP—secreted frizzled-related proteins; TCF—T cell factor; β-TrCP—beta- repeats-containing proteins; WIF1—WNT inhibitory factor 1; WISE—WNT modulator in surface ; Ub; ubiquitin; ZNRF3—zinc and ring finger protein 3. Int. J. Mol. Sci. 2020, 21, x FOR PEER REVIEW 3 of 32

related protein; MAK—metastasis associated kinase; PAR1—protease-activated receptor 1; PKC—protein Int. J. Mol. Sci. 2020kinase, 21 ,C 4852; PYGO—pygopus; RNF43—ring finger protein 43; sFRP—secreted frizzled-related proteins; TCF— 3 of 31 T cell factor; β-TrCP—beta-transducin repeats-containing proteins; WIF1—WNT inhibitory factor 1; WISE— WNT modulator in surface ectoderm; Ub; ubiquitin; ZNRF3—zinc and ring finger protein 3.

Figure 2. An overview of non-canonical WNT-signaling pathways: (A) WNT/planar cell polarity-signaling Figure 2. pathwayAn overview (PCP) is initiated of non-canonical by WNT binding WNT-signaling to FZD and ROR, pathways: then DVL is recruited (A) WNT and / DVLplanar-Daam cell-1 polarity- signaling pathwaycomplex is activated, (PCP) isfollowed initiated by JNK by and WNT ROCK binding activation and to FZDcytoskeletal and rearrangement; ROR, then DVL(B) WNT/Ca is recruited2+- and DVL-Daam-1signaling complex pathway is activated, is initiatedfollowed by WNT bybinding JNK to and FZD ROCK and ROR, activation with further and cytoskeletal G-protein triggered rearrangement ; phospholipase2+ C activation leading to intracellular calcium fluxes and downstream calcium (B) WNT/Cadependent-signaling responses. pathway AP-1—activator is initiated protein by 1; WNT CaMKII binding—Ca2+/calmodulin to FZD and dependent ROR, kinase with II; further CaN— G-protein triggered phospholipasecalcineurin; CDC42 C activation—cell division leading cycle 42; to phospholipaseDAG—diacylglycerol; C intracellular DAAM1—DVL calcium associated fluxes activator and downstream of calcium dependentmorphogenesis responses.; DVL—disheveled; AP-1—activator FZD—frizzled; protein JNK—c 1;-Jun CaMKII—Ca2 N-terminal ;+/calmodulin NLK—nemo like dependent kinase; kinase NFAT—nuclear factor of activated T-cells; PIP2—phosphatidylinositol (4,5)-biphosphates; PKC—protein II; CaN—; CDC42—cell division cycle 42; DAG—diacylglycerol; DAAM1—DVL associated kinase C; PLC—phospholipase C; RAC—Ras-related C3 botulinum toxin ; RHO—Ras homolog activator ofgene ; family; ROCK— DVL—disheveled;Rho-associated kinase; FZD—frizzled; ROR—RAR-related JNK—c-Jun ; N-terminal TAK1— kinases;transforming NLK—nemo like kinase;growth NFAT—nuclear factor beta-activated factor kinase of 1. activated T-cells; PIP2—phosphatidylinositol (4,5)-biphosphates;

PKC—protein1.2. β-Catenin kinase-Dependent C; PLC—phospholipase(Canonical) WNT Signaling C; RAC—Ras-related C3 botulinum toxin substrate; RHO—Ras homolog gene family; ROCK—Rho-associated kinase; ROR—RAR-related orphan receptor; In resting cells, in the absence of activating signals (Figure 1A), the level of β-catenin is low, which is TAK1—transformingachieved by the cytoplasmicgrowth factor ‘destruction beta-activated complex’ kinase that 1. consists of axis inhibition protein 1 (AXIN1), adenomatosis polyposis coli (APC), glycogen synthase kinase 3β (GSK3β) and casein kinase 1α (CK1α) 1.2. β-Catenin-Dependent[7,21–23]. AXIN1 is (Canonical)the central scaffold WNT protein, Signaling which directly interacts with all other core components of In resting cells, in the absence of activating signals (Figure1A), the level of β-catenin is low, which is achieved by the cytoplasmic ‘destruction complex’ that consists of axis inhibition protein 1 (AXIN1), adenomatosis polyposis coli (APC), glycogen synthase kinase 3β (GSK3β) and casein kinase 1α (CK1α)[7,21–23]. AXIN1 is the central scaffold protein, which directly interacts with all other core components of the destruction complex [24]. It is a concentration-limiting protein [25], and its cellular level is stabilized by SUMOylation [26] and decreased by degradation involving activated low-density lipoprotein receptor related protein 5/6 (LRP5/6) [27] and [28]. Affinity of AXIN1 to β-catenin is increased by GSK3β-mediated [29,30]. In the destruction complex, β-catenin is first phosphorylated by CK1α at Ser45, then by a / kinase GSK3β at Ser 33, Ser 37 and Thr41. Interaction of GSK3β with β-catenin is facilitated by AXIN1 and APC [29,31]. F-box containing protein E3 ubiquitin ligases such as β-transducin repeat-containing protein (β-TrCP) marks β-catenin for ubiquitination and proteasomal degradation [4,22,23,32]. Upon binding of WNT proteins to the seven-pass transmembrane frizzled (FZD) receptor and single pass transmembrane receptors LRP5 and LRP6, the initiation of canonical signaling pathway occurs, leading to β-catenin stabilization (Figure1B) [ 7,32]. A characteristic feature of FZD is the cysteine-rich Int. J. Mol. Sci. 2020, 21, 4852 4 of 31 domain, which is the primary module for binding of WNT ligands [4,13]. The mutual interaction of WNT ligands with both FZD and LRP5/6 is necessary for canonical pathway activation that inhibits β-catenin proteasomal degradation [7,22,32]. It is preceded by a series of events, and several models have been created [33] to show the sequence of events leading to activation of β-catenin. Regardless of the model, disheveled (DVL) that directly interacts with FZD is concomitantly phosphorylated by several protein kinases such as protease-activated receptor 1 (PAR1), casein kinase 1ε (CK1ε), metastasis associated kinase (MAK) and protein kinase C (PKC) [14,34]. Activated DVL detaches AXIN1 from the destruction complex and the released AXIN1 binds to phosphorylated LRP5/6 [7,32]. Phosphorylation of LRP5 and LRP6 occurs in Pro-Pro-Pro(SerTrp)Pro (PPP(S/T)P) motifs and is triggered by GSK3β and CK1γ [23,34]. β-catenin can be dephosphorylated by protein 2A (PP2A) [35]. WNT/β-catenin-signaling can be potentiated by the leucine-rich repeat-containing G-protein coupled receptor 5/a roof plate-specific spondin (LGR5/RSPO) complex, which acts in cooperation with receptors FZD/LRP5/6 [36–38]. The LRG5/RSPO complex promotes WNT-signaling through the neutralization ring finger protein 43 (RNF43) and zinc and ring finger protein 3 (ZNRF3), the transmembrane E3 ligases that serve as a part of a negative feedback loop [39]. All these processes lead to the accumulation of stable, unphosphorylated β-catenin in the , followed by its translocation to the nucleus. β-catenin binds to nucleoporins (NUPs) and builds the nuclear pore complex (NPC). It binds NUP358 on the cytoplasmic side, NUP62 in a central channel and NUP98 together with NUP153 on a nuclear end [40]. In the nucleus, after displacing the transcriptional repressor Groucho, β-catenin can interact with several proteins, e.g., T cell factor/lymphoid enhancer-binding factor 1 (TCF/LEF), brahma-related gene-1 (BRG-1), TATA-binding protein, CREB-binding protein/its homolog p300 (CBP/p300), c-JUN, SWItch/sucrose non-fermentable chromatin-remodeling complex (SWI/SNF) and B-cell CLL/lymphoma 9 protein (BCL-9; BCL9L), which links the N terminal part of β-catenin with pygopus (PYGO) [34,41]. Then, β-catenin serves as a transcriptional regulator of the expression of WNT target genes [7,21,32,34]. These genes encode the following groups of proteins: (a) regulators of proliferation, e.g., vascular endothelial (VEGF), fibroblast growth factor (FGF), c-JUN, FOS-related antigen 1 (FRA1); (b) regulators of the canonical WNT pathway e.g., WNT1-inducible-signaling pathway protein 1 (WISP1), AXIN, Dickkopf-1 (DKK1), TCF, LEF1; (c) matrix metalloproteinases and some components of extracellular matrix; (d) cadherins; (e) lineage-specific proteins such as microphthalmia-associated factor (MITF), which modulates several functions in melanocytes and melanoma [42]. It should be noted that the transcriptional role of β-catenin extends beyond the TCF/LEF as β-catenin may be a partner of other transcription factors, e.g., sex-determining region Y (SRY) box-containing factors (SOX), mothers against decapentaplegic homolog (SMAD), octamer-binding 4 (OCT4) and forkhead box class O family member proteins (FOXOs) [43]. WNT/β-catenin-signaling can be modulated by several antagonists at the ligand and receptor level, e.g., DKK proteins, secreted frizzled-related proteins (sFRPs), WNT inhibitory factor 1 (WIF1), WNT modulator in surface ectoderm (WISE), Kremen (KRM) and protein (CER1) [14,16,44]. They interact with WNTs and their receptors, causing the inhibition of the WNT/β-catenin pathway [16]. Moreover, the activity of WNTs is also modulated by a highly conserved feedback antagonist NOTUM, acting as a deacylase that removes a palmitate moiety from WNTs leading to their inactivation [4]. There are several negative feedback mechanisms that can limit WNT-signaling, including WNT target genes such as AXIN1, AXIN2, DKK1 and SFRP [45,46]. The β-catenin-dependent WNT-signaling cascade (Figure1) regulates a wide range of biologic processes comprising both developmental processes during embryogenesis as well as those during tissue homeostasis and regeneration. Canonical WNT-signaling is involved in the regulation of cell proliferation and differentiation and maintenance of stem cells. β-catenin, the central component of WNT/β-catenin pathway is a multifunctional protein that can either bind to cadherin that is an integral part of the or act as transcriptional [42,47]. β-catenin is expressed both in melanocytes and epithelium, however, the amount of β-catenin at the cell surface is less abundant in melanocytes than in epithelial cells [48]. β-catenin (781 amino acids) is encoded by CTNNB1 and Int. J. Mol. Sci. 2020, 21, 4852 5 of 31 comprises a flexible N-terminal domain (NTD; ~150 aa), central armadillo (ARM) repeat domain (12 copies, 550 aa) and a C-terminal transactivation domain (CTD; ~100 aa) [23]. While NTD and CTD may be flexible, the central region forms a rigid scaffold that is responsible for the interaction of β-catenin with its binding partners [49] such as cytoskeletal proteins α-catenin, IQ motif containing GTPase activating protein 1(IQGAP1) [50], E-cadherin and N-cadherin [51], a conserved nuclear protein, named chibby, transcriptional regulators TCF/LEF, inhibitor of β-catenin and TCF4 (ICAT) and proteins forming the ‘destruction complex’.

1.3. Non-Canonical WNT Signaling β-catenin-independent pathways comprise: WNT/planar cell polarity-signaling pathway (PCP) (Figure2A) and WNT /Ca2+-signaling pathway (Figure2B). The WNT /PCP-signaling pathway maintains planar cell polarity, as it is involved in regulation of modification of actin cytoskeleton structures and cell motility. It is activated by binding of WNT5A, WNT7A and WNT11 ligands to non-canonical FZD receptors along with kinase co-receptors: protein 7 (PTK7), RAR-related orphan receptor (ROR) and receptor like tyrosine kinase (RYK) [52]. WNT5A is a key regulator of non-canonical WNT-signaling, however, it can play diverse roles in different types of cells, including tumor cells. Different roles of WNT5A, especially in various cancers, are partially due to the existence of two isoforms of WNT5A, WNT5A-long and WNT5-short [53]. Activation of DVL and further formation of the disheveled associated activator of morphogenesis 1 (DVL-Daam-1) complex activates the RHO GTPase that leads to activation of the RHO-associated kinase (ROCK) followed by modification of actin cytoskeleton and cytoskeletal rearrangement. DVL also activates the RAC GTPase that stimulates c-Jun N-terminal kinases (JNK) activity, which in turn positively regulates activator protein 1 (AP-1)-dependent genes. This network is involved in the modification of actin cytoskeleton structures influencing the polarization and motility of cells [17,54]. The WNT/Ca2+-signaling pathway is associated with the release of Ca2+ from intracellular stores. Interaction of WNT ligands with FZD activates phospholipase C (PLC) that hydrolyzes phosphatidylinositol (4,5)-biphosphates (PIP2) to inositol (1,4,5)-triphosphates (IP3) and diacylglycerol (DAG). DAG activates PKC kinase that in turn activates the small GTPase CDC42, while IP3 induces the release of Ca2+ ions from intracellular depots. Release of Ca2+ activates Ca2+/calmodulin dependent kinase II (CaMKII) and calcineurin (CaN). CaMKII phosphorylates TGFβ-activated kinase 1 (TAK1), which induces Nemo-like kinase (NLK) activation, which in turn inhibits transcriptional activity of canonical WNT-signaling. CaN via dephosphorylation activates nuclear factor of activated T-cells (NFAT) that translocates to the nucleus and regulates the expression of target genes. WNT/Ca2+ pathway activation plays an important role in the regulation of cell motility and cytoskeleton organization [17,54,55].

2. WNT Signaling in Cancer As demonstrated in numerous studies, the aberrant activation of WNT-signaling contributes to malignant cell transformation and neoplastic proliferation with further metastatic dissemination and resistance to treatment [43,56]. Many surface markers of cancer stem cells (CSCs) (CD44, CD24, CD133, LGR5/GPR49, ABC cassette genes, EpCAM) are direct targets of the WNT pathway. CSCs provide the long-term maintenance of the tumor and contribute to poor clinical outcome of therapies [43]. Different genetic alterations can cause the inhibition of proteasomal degradation of β-catenin, resulting in the hyperactivation of canonical WNT-signaling and enhanced nuclear β-catenin accumulation. Genetic and epigenetic alterations affecting constituents of WNT pathways are tissue specific and they differ in between cancers [56]. Apart from the most frequent mutations of APC in and CTNNB1 in hepatocellular carcinoma, deregulations of several extracellular modulators of WNT-signaling e.g., DKKs, sFRPs and WIF1 also contribute to cancer development. These proteins antagonize canonical WNT-signaling by binding to LRP5/6 or inhibit the interaction between WNTs and their receptors [6]. Moreover, vacuolar H+-ATPase (v-ATPase), an electrogenic H+ Int. J. Mol. Sci. 2020, 21, 4852 6 of 31 transporter required for WNT-signaling activation may also trigger abnormal WNT/β-catenin-signaling and contribute to WNT-signaling-dependent tumorigenesis. High expression of v-ATPase subunits has been observed in colorectal, prostate, breast, ovarian and cells [23]. Furthermore, it has also been demonstrated that β-catenin modulates the cancer microenvironment, participating in creating the niche for cancer progression [57]. The pre-metastatic niche permits both the implantation of tumor cells into distant organs as well as their survival [58–61]. However, high levels of nuclear β-catenin do not always indicate poor prognosis [57]. For that reason, it is necessary to consider the cell type-specific background in order to assess and understand the cellular outcome of aberrations in WNT-signaling [62].

3. WNT Signaling in Melanoma

3.1. Genetic and Epigenetic Alterations Deregulations in the canonical WNT-signaling in cancer may result from diverse mutations and epigenetic mechanisms. Mutations in genes encoding distinct elements of the WNT pathway can cause (i) loss of function of the destruction complex, (ii) increase in nuclear localization of β-catenin resulting in β-catenin-mediated transactivation of several genes. Mutations are mostly detected in genes encoding components of the destruction complex such as APC and AXIN, but the gene encoding β-catenin, CTNNB1, is also frequently mutated in selected cancer types. High frequency of mutations causing hyperactivation of WNT-signaling can be detected for example in colorectal cancer [63,64] and mutations in APC leading to APC loss of function have been found in about 70% of cases [65–67]. In melanoma, frequencies of mutations in APC, AXIN1 and CTNNB1 are low, reaching according to cbioportal.org 10%, 2.9% and 5.9%, respectively (Table1), however, a significant interstudy variability exists. An early study reported mutations in CTNNB1 in six of 26 melanoma cell lines, and all these mutations affect phosphorylation of β-catenin rending it resistant to proteasomal degradation [68]. Several later studies have shown much lower frequencies of mutations in CTNNB1 and APC, which put into question the importance of a genetic component in aberrant activity of β-catenin in melanoma [69–73]. For example, only 1 of 65 primary harbored mutations of CTNNB1, and one third of cases showed nuclear accumulation of β-catenin [69]. In another study, while a CTNNB1 missense and a truncating APC mutation were reported only in one out of forty cell lines, it has been demonstrated that hypermethylation of APC was present in about 15% of melanoma biopsies and cell lines suggesting transcriptional silencing [74]. This epigenetic regulation was, however, not accompanied with increased WNT-signaling probably due to residual activity of APC. Epigenetic regulation of WNT/β-catenin antagonists such as DKKs, WIF1 and sFRP2 contributes substantially to activation of β-catenin. DKK1, DKK2 and DKK3 downregulation has been observed in melanoma cell lines and tissue samples, however, in contrast to other cancer types promoter hypermethylation is responsible only for downregulation of DKK2 [75]. Promoter methylation of WIF1 has been shown as leading to WIF1 silencing [76,77]. Luo et al. have found that methylation of the sFRP2 promoter results in a significant decrease of sFRP2 in patient melanoma samples and melanoma cell lines compared with the paired adjacent non-tumor tissue and non-transformed melanocytes, respectively [78]. Int. J. Mol. Sci. 2020, 21, 4852 7 of 31

Table 1. Frequency of mutations of genes encoding elements of the canonical WNT-signaling pathway detected in melanoma.

Melanoma Gene Literature Data (cbioportal.org; 1284 Cases) 15% [74] APC 11% [79] 10.0% 2.7% [73] AXIN1 – 2.9% AXIN2 11% [79] 1.3% BCL9 – 5.7% 23% [68] 4.6% [71] 3.3% [71] CTNNB1 11% [79] 5.9% 1.6% [72] 1.5% [69] 5.4% [73] 3.2% [70] 3.7% [70] 8.1% [80] FBXW7 3.4% 8.3% [81] GSK3β – 1.2% SFRP – 1.7% WIF1 – 2.8%

3.2. Canonical Signaling WNT-signaling plays an important function in skin. WNT/β-catenin-signaling guides the migration of neural crest cells, multipotent precursor cells and drives them toward a melanocyte fate [82–84], including terminal differentiation of melanoblasts to melanocytes [83]. WNT/β-catenin-signaling is also responsible for the maintenance of homeostasis between melanocytes and keratinocytes in normal epidermis [44,54]. The activation of MITF, a lineage-specific transcription factor, by the canonical WNT-signaling plays an important role in melanocytes [85]. It has been observed that the fate of neural crest cells depends on MITF, as cells with low MITF level differentiate toward e.g., neurons, glial cells or cartilage, whereas cells with high MITF give rise to melanocytes [84]. In the context of melanocyte lineage, β-catenin not only controls expression of MITF, but also modulates its function via direct protein–protein interaction [86]. As melanoma cells utilize WNT/β-catenin-signaling for transformation and proliferation, whereas non-canonical signaling for metastasis [87], β-catenin does not fully comply with the definition of an oncogene in this cancer [88]. The exact role of WNT-signaling in melanoma initiation and progression remains highly controversial to date despite extensive studies. While an increasing level of nuclear β-catenin during cancer progression is characteristic for several cancer types including colorectal cancer and hepatocellular carcinoma [89–92], the level of nuclear β-catenin is decreased in melanoma during disease progression [62,93–97]. However, opposite results have been published as well [98–100]. β-catenin hyperactivation in melanoma is rarely caused by mutations of CTNNB1 [101]. β-catenin has been shown to play a critical role in the early stages of melanocyte transformation [102,103]. The initiation of melanoma includes enhanced proliferation of cells together with suppression of Int. J. Mol. Sci. 2020, 21, 4852 8 of 31 senescence leading to abnormal growth of melanocytes [104]. Senescence is mediated by two main tumor suppressor pathways: INK4α/RB and ARF/ [105]. The INK4α-ARF (CDKN2α) encodes two proteins: p16INK4a and p14(p19)ARF, regulating RB1 and p53 pathways, respectively. Inactivation of the genetic locus INK4α-ARF is important in overcoming the senescence barrier to oncogenesis [104]. In a variety of cancers, this locus is inactivated by mutations, whereas it has also been found that p16INK4a expression can be silenced by activated β-catenin [103]. Delmas et al. observed the ability of β-catenin to bypass senescence by silencing the p16INK4a through a conserved TCF/LEF site in its promoter [103]. Activation of β-catenin in melanoma is a consequence of binding of WNT1 and WNT3A to their receptors FZD1/7 and co-receptors LRP5/6 and ROR1. These two ligands are crucial for bypassing melanocyte senescence and inducing transformation of melanocytes [103,106]. While benign nevi have been found to be positive for nuclear β-catenin, loss of nuclear β-catenin during melanoma progression to metastases has been reported [62,94,107]. The importance of canonical WNT-signaling in melanoma initiation is mostly associated with β-catenin ability to regulate the expression of a wide range of genes of the melanocyte lineage, and its involvement in regulation of proliferation is most likely related to the activation of MITF expression [42,44,62,108–110]. The rheostat model and phenotype switching model of MITF function in melanoma [111–113] suggest that a very low MITF level or the absence of MITF results in cell senescence, a low level is characteristic for invasive/dedifferentiated melanoma cells, whereas a high level of MITF pushes cells first toward a highly proliferative phenotype and finally to differentiation. Therefore, MITF is considered to be a master regulator of ‘phenotype switching’ between proliferative and invasive states, contributing to the high plasticity of melanoma cells in response to changes in the tumor microenvironment, including response to treatment [82,114–117]. Moreover, MITF plays an important pro-survival role in melanoma cells [118]. Recently, a differentiation model of melanoma has been proposed, in which the level of MITF is one of crucial factors determining subtypes of melanoma exerting differential vulnerability to drug-induced stress [119]. In view of the correlation between β-catenin and MITF expression, WNT-signaling also affects melanoma plasticity [120]. In actively proliferating melanoma cells, nuclear β-catenin/LEF1 triggers the expression of MITF that in turn activates transcription of several genes, including genes encoding regulators , and dependent kinase 2 (CDK2, pigmentation-related proteins such tyrosinase, dopachrome tautomerase (DCT) and Melan-A and pro-survival factors e.g., BCL2 and BCL2A1 [42,118,121]. Active β-catenin-signaling leads to increased differentiation in benign lesions [62,117] as it disrupts the regulatory balance between PAX3, SOX10 and MITF towards terminal differentiation [83]. It has been found that β-catenin mediates the activation of POU domain of transcription factor BRN2 that plays a complex role in regulation of MITF [83,101]. BRN2 tends to be highly expressed in melanomas, which suggests that it can be a positive regulator of melanoma survival and proliferation [83]. It has been reported that depending on the cellular context, BRN2 can transcribe or repress MITF. It can drive MITF-mediated proliferation in the presence of oncogenic BRAF, whereas in the absence of mutated BRAF it can represses MITF promoting invasion [101]. Moreover, the proliferative MITFhigh subpopulation of melanoma cells producing melanoma antigen recognized by T cells 1 (MART1) are less invasive, whereas when this differentiation antigen is lost melanoma cells acquire highly invasive properties [122]. Differential behavior of β-catenin in epithelial cancers and melanoma is associated with opposing features of epithelial cells and melanocytes [99]. Therefore, it has been strongly emphasized that these diverse effects of β-catenin in melanoma cells and epithelial-derived cancers may be connected with the activity of MITF [62,99]. Arozarena et al. have shown the correlation between the expression of β-catenin and MITF both in cell lines and melanoma biopsies [62]. When low β-catenin and highly invasive cells were treated with forskolin, an agent upregulating MITF, the complete loss of membrane blebs and F-actin cortex contraction was observed. In line with this, melanoma cells with high β-catenin and high MITF levels have been characterized with an elongated filopodia phenotype and MITF reduction significantly increased rounded blebbing cells [62]. The round shape morphology is regulated by Int. J. Mol. Sci. 2020, 21, 4852 9 of 31 phosphorylation of RHO/ROCK-mediated myosin light chain (MLC) that induces the contraction of the cortical actin meshwork, regulating bleb retraction of cells. Aberrant expression of WNT/β-catenin antagonists (e.g., DKKs, WIF1, sFRPs) is common in melanoma and is associated with elevated β-catenin level. Downregulation of DKKs has been detected in melanoma cell lines and tissue samples [75]. Reduced level of WIF1 has been found to contribute to constitutive activation of canonical WNT-signaling in melanoma cells, and WIF1 overexpression resulted in downregulation of WNT-signaling and suppression of melanoma cell proliferation [76]. Differences in WIF1 expression have been observed between its level in the primary tumor and sentinel lymph node (SLN) metastases [77]. Similarly, decreased expression of sFRPs, leading to upregulated WNT/β-catenin-signaling is also common in melanoma, and sFRP1 has been shown as an appealing candidate for a tumor suppressor [123]. Reduction of sFRP2 enhances the canonical WNT-signaling, whereas demethylation of its promoter inhibits the nuclear retention of β-catenin in melanoma cells and suppresses invasion [78]. Kaur et al. have found that sFRP2 is expressed by aged fibroblasts that play an important role in melanoma progression and this expression is associated with melanoma metastasis, therapy resistance and poor outcomes in elderly patients [82]. sFRP2 decreases β-catenin, MITF and apurinic/apyrimidinic endonuclease 1 (APE1). APE1 is the redox effector involved in DNA repair, so its decrease can attenuate the response of melanoma cells to DNA damage induced by , making melanoma cells more resistant to [82].

3.3. Non-Canonical WNT Signaling Non-canonical WNT-signaling, which is activated during melanoma progression, suppresses the WNT/β-catenin-signaling [107,124]. The interaction of canonical and non-canonical WNT-signaling in melanoma is presented in Figure3. A lower level of nuclear β-catenin was detected in metastases than in primary melanoma in 343-melanoma samples [107]. Immunohistochemical analyses have revealed that most metastatic melanoma exhibited strong WNT5A staining comparing to benign tumors and a high WNT5A level in metastatic cutaneous melanoma is associated with poor outcome [125]. Therefore, it has been suggested that lack of nuclear β-catenin and high level of cytoplasmic WNT5A can be indicatives of unfavorable prognosis for melanoma patients [62,107,125]. Furthermore, inducible overexpression of WNT5A in melanoma cells exhibiting low metastatic activity results in enhanced invasiveness [126], whereas siRNA-mediated knockdown of ROR2, the WNT5A receptor, reduced the frequency and severity of metastases in mice [127]. Pro-metastatic effects of WNT5A have been associated with aerobic glycolysis promoted by WNT5A in melanoma cells [128]. WNT5A-mediated Ca2+-dependent release of exosomes by melanoma cells has been reported [129]. Exosomes are the smallest type of extracellular vesicles (30–180 nm in diameter), whose role in melanoma has been extensively discussed recently [61]. Using recombinant WNT5A in melanoma cells expressing low endogenous WNT5A induces a rapid release of exosomes containing the immunomodulatory 6 (IL-6) and the pro-angiogenic factors interleukin 8 (IL-8), VEGF and matrix metalloproteinase-2 (MMP2), whereas WNT5A depletion in melanoma cells expressing high endogenous WNT5A causes the reduction of IL-6, IL-8, VEGF and MMP2. Release of immunomodulatory and pro-angiogenic molecules enhances aggressiveness of melanoma cells and their capacity to metastasize [129]. Furthermore, it has been reported that WNT5A increases , which mediates resistance to various anticancer drugs. Knocking down autophagy-related gene 5 (ATG5) in WNT5Ahigh melanoma cells caused a reduction of the WNT5A level and induction of β-catenin [130]. Int. J. Mol. Sci. 2020, 21, 4852 10 of 31 Int. J. Mol. Sci. 2020, 21, x FOR PEER REVIEW 10 of 32

Figure 3. Proposed model of the crosstalk between canonical and non-canonical WNT-signaling in Figure 3. Proposed model of the crosstalk between canonical and non-canonical WNT-signaling melanoma. in In melanoma. the canonical In the WNT canonical pathway, WNT pathway, WNT–FZD/LRP5/6 WNT–FZD/LRP5 interaction/6 interaction initiates initiates β-βcatenin-catenin dependent signaling. βdependent-catenin translocates signaling. β-catenin to the translocatesnucleus to to drive the nucleus the transcriptio to drive then transcription of target genes. of target This genes. is critical for early steps Thisof transformation is critical for early when steps melanocytes of transformation bypass when senescence melanocytes and bypass start senescence to proliferate, and start thus to promoting first the radialproliferate, then vertical thus promoting growth firstof melanoma. the radial then An vertical increase growth of WNT5A of melanoma. that Anactivates increase non of WNT5A-canonical WNT- that activates non-canonical WNT-signaling inhibits β-catenin-signaling and enhances the invasiveness signaling inhibits β-catenin-signaling and enhances the invasiveness of melanoma cells crucial for metastatic of melanoma cells crucial for metastatic spreading of melanoma. Green and red arrows indicate increase spreading ofand melanoma. decrease, respectively. Green and red arrows indicate increase and decrease, respectively.

WNT5A canWNT5A either can inhibit either inhibit or activate or activate canonical canonical WNT-signaling WNT-signaling in diverse in diverse ways depending ways depending on the on the receptor contextreceptor [131]. context Several [131]. Several of these of these mechanisms mechanisms have have been been investigated investigated in melanoma in melanoma and the results and the results revealed that WNT5A exerts a dichotomous role in melanoma (Figure3). It can either stabilize β-catenin revealed thatvia WNT5A adenosine exerts diphosphate a dichotomous (ADP)-ribosylation role in factor melanoma 6 (ARF6) or(Figure suppress 3). the It WNTcan either/β-catenin-signaling stabilize β-catenin via adenosine diphosphateeither by binding (ADP) to FZD-ribosylation receptors or infactor a seven 6 (ARF6) in absentia or suppress homolog 2 the (SIAH2)-dependent WNT/β-catenin manner.-signaling either by bindingBinding to FZD of receptors WNT5A to or FZD4 in a/LRP6 seven receptor in absentia complex homolog activates ARF62 (SIAH2) that in-dependent turn releases manner.β-catenin Binding of WNT5A tofrom FZD4/LRP6 N-cadherin, receptor which stimulatescomplex theactivates shuttling ARF6 of β -cateninthat in betweenturn releases the membrane, β-catenin cytoplasm from N-cadherin, which stimulatesand nucleus the shuttling and enhances of ββ--catenin-dependentcatenin between the transcription membrane, [132 ].cytoplasm WNT5A that and signals nucleus through and enhances FXD2, FZD5 and ROR2 activates PKC/Ca2+-signaling [87]. Calpain-1 (CAPN) mediates the cleavage of β-catenin-dependent transcription [132]. WNT5A that signals through FXD2, FZD5 and ROR2 activates filamin A, promoting the remodeling of cytoskeleton and enhancing the motility of melanoma cells, 2+ PKC/Ca -signalingwhereas CAMKII [87]. Calpain inhibits-1β -catenin(CAPN) activity mediates [87]. the PKC cleavage activates signalof filamin transducer A, promoting and activator the of remodeling of cytoskeleton and enhancing the motility of melanoma cells, whereas CAMKII inhibits β-catenin activity [87]. PKC activates signal transducer and activator of transcription 3 (STAT3) leading to inhibition of MITF expression with concomitant downregulation of melanocytic differentiation antigens (Melan A (MART1), DCT and gp100), thus promoting the metastatic phenotype [87]. Furthermore, PKC influences the motility of melanoma cells through the inhibition of metastasis repressor KISS-1 and E-cadherin and upregulation of metastasis-associated molecules, CD44 and SNAIL, resulting in transition to an invasive phenotype [87,133]. WNT5A can compete with WNT3A for binding to FZD2, and in this way LPR6 phosphorylation and β-catenin accumulation can be inhibited [122,134].

Int. J. Mol. Sci. 2020, 21, 4852 11 of 31 transcription 3 (STAT3) leading to inhibition of MITF expression with concomitant downregulation of melanocytic differentiation antigens (Melan A (MART1), DCT and gp100), thus promoting the metastatic phenotype [87]. Furthermore, PKC influences the motility of melanoma cells through the inhibition of metastasis repressor KISS-1 and E-cadherin and upregulation of metastasis-associated molecules, CD44 and SNAIL, resulting in transition to an invasive phenotype [87,133]. WNT5A can compete with WNT3A for binding to FZD2, and in this way LPR6 phosphorylation and β-catenin accumulation can be inhibited [122,134]. WNT5A can act through SIAH2 via the GSK3-β-independent pathway to promote degradation of β-catenin [135–137]. This mechanism of switching from WNT/β-catenin-signaling to non-canonical WNT-signaling has been associated with resistance to targeted therapy [136]. A high level of WNT5A promotes the resistance to vemurafenib, highlighting the opposing roles of different WNT pathways in melanoma [87]. Melanoma cells exerting a high level of WNT5A when exposed to vemurafenib treatment or other stress, developed a sort of drug resistant state (pseudosenescence), still retaining invasive capacity and the ability to form metastases. Furthermore, the pseudosenescent WNT5A-high cells were capable to express factors similar to those secreted by senescent fibroblasts and were positive for senescence markers, e.g., β-galactosidase, Src 2 domain containing F (SHF) and methylated of histone H3 on lysine 9 (H3K9Me). This WNT5A-mediated senescence-like response may be the mechanism that allows the tumor to evade therapy by undergoing growth arrest [122]. FZD3 has been found to be associated with β-catenin-independent-signaling and upregulation of this receptor is associated with melanoma progression and a reduced patient survival, whereas down-regulation of FZD3 suppresses growth and metastasis of melanoma [138]. Overexpressed FZD3 has been detected in 20% of melanoma patients whose tumors were deprived of infiltrating T cells indicating the importance of this receptor in immune evasion [139]. Moreover, it has been observed that FZD3 effectively modulates the activity of the -activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) pathway that is critical for melanoma maintenance. Therefore, FZD3 blocking agents may enhance the efficacy of melanoma treatment when used in combination with available kinase inhibitors and immunotherapy [138,139].

4. Crosstalk between WNT Signaling and Key Oncogenic Pathways Involved in Melanoma Development, Progression and Response to Treatment The complex interactions existing between diverse signaling pathways create the opportunity for a cancer cell to compensate for a blockade of a single pathway. The crosstalk consisting of multiple interactions and feedback loops is crucial for the cellular response to microenvironmental changes, including treatment. WNT-signaling is an important part of the crosstalk between key oncogenic pathways involved in melanoma development, progression and response to treatment. Elements of the WNT-signaling pathways both depend on and regulate activities of diverse pathways such as MAPK/ERK and phosphatidylinositol-3-kinase (PI3K)/AKT as well as transcriptional regulators including p53 and MITF. Figure4 shows main signal transduction pathways and transcriptional regulators that interact with the WNT-signaling pathways in melanoma. Int. J. Mol. Sci. 2020, 21, 4852 12 of 31 Int. J. Mol. Sci. 2020, 21, x FOR PEER REVIEW 12 of 32

FigureFigure 4. 4.MainMain signal signal transduction transduction pathways pathways and and transcriptional transcriptional regulators regulators that thatinteract interact with with the WNT the - signalingWNT-signaling pathways pathways in melanoma. in melanoma.Figure shows Figurethe possible shows crosstalk the possible between crosstalk different betweenpathways, di however,fferent somepathways, of the interactions however, some are ofgenetic the interactions context-, disease are genetic stag context-,e- or treatment disease-dependent. stage- or treatment-dependent. See the text for more details. APC—adenomatosis polyposis coli; AXIN—axis inhibition protein; BCL—B-cell CLL/lymphoma See the text for more details. APC—adenomatosis polyposis coli; AXIN—axis inhibition protein; protein; BRG-1—brahma-related gene-1; CBP—(CREB)-binding protein; CK1α—casein kinase 1α; COX-2— BCL—B-cell CLL/lymphoma protein; BRG-1—brahma-related gene-1; CBP—(CREB)-binding protein; cyclooxygenase 2; DVL—disheveled; FZD—frizzled receptor; GSK3β—glycogen synthase kinase 3β; CK1α—casein kinase 1α; COX-2—cyclooxygenase 2; DVL—disheveled; FZD—frizzled receptor; iASPP—inhibitor of apoptosis-stimulating protein of p53; LEF—lymphoid enhancer-binding factor 1; LRP— GSK3β—glycogen synthase kinase 3β; iASPP—inhibitor of apoptosis-stimulating protein of low-density lipoprotein receptor related protein; MITF—microphthalmia-associated transcription factor; p53; LEF—lymphoid enhancer-binding factor 1; LRP—low-density lipoprotein receptor related PTEN—phosphatase and tensin homolog deleted on ten; PYGO—pygopus; mTOR— protein; MITF—microphthalmia-associated transcription factor; PTEN—phosphatase and tensin mammalian target of rapamycin; PI3K— phosphatidylinositol-3-kinase; RAS—Rat sarcoma.; ROR—RAR- homolog deleted on chromosome ten; PYGO—pygopus; mTOR—mammalian target of rapamycin; related orphan receptor; RTK——SIAH2—seven in absentia homolog 2. PI3K—phosphatidylinositol-3-kinase; RAS—Rat sarcoma.; ROR—RAR-related orphan receptor; RTK—receptor tyrosine kinase—SIAH2—seven in absentia homolog 2. WNT-signaling can interact with MAPK/ERK and PI3K/AKT-signaling in diverse ways. Moreover, growingWNT-signaling evidence indicates can interact that this with crosstalk MAPK/ ERKin melanoma and PI3K depends/AKT-signaling on the ingenetic diverse context ways. and Moreover, several mechanismsgrowing evidence are considered indicates as that tumor this stage crosstalk-dependent in melanoma or unique depends for melanoma. on the genetic In an context early andstudy, several it was shown that WNT/β-catenin and MAPK/ERK pathways synergized in order to induce melanoma without mechanisms are considered as tumor stage-dependent or unique for melanoma. In an early study, it was the need for p16INK4a mutations and β-catenin could cooperate with N-RAS [103]. Next, it was found that shown that WNT/β-catenin and MAPK/ERK pathways synergized in order to induce melanoma without inhibition of BRAFV600 enhanced the activity of β-catenin in the presence of WNT ligands [140]. This the need for p16INK4a mutations and β-catenin could cooperate with N-RAS [103]. Next, it was found combination of BRAF inhibition and WNT/β-catenin activation was accompanied by AXIN1 degradation that inhibition of BRAFV600 enhanced the activity of β-catenin in the presence of WNT ligands [140]. and inhibition of GSK3β, whereas hyperactivated MAPK/ERK-signaling stabilized AXIN1 that inhibited This combination of BRAF inhibition and WNT/β-catenin activation was accompanied by AXIN1 WNT-signaling in melanoma [140]. This negative crosstalk between MAPK/ERK and WNT/β-catenin- degradation and inhibition of GSK3β, whereas hyperactivated MAPK/ERK-signaling stabilized AXIN1 signaling was considered as melanoma specific since in colon cancer and many other cancers, activation of that inhibited WNT-signaling in melanoma [140]. This negative crosstalk between MAPK/ERK and

Int. J. Mol. Sci. 2020, 21, 4852 13 of 31

WNT/β-catenin-signaling was considered as melanoma specific since in colon cancer and many other cancers, activation of WNT/β-catenin-signaling leads to the stabilization of both β-catenin and RAS and activation of MAPK-signaling [141,142]. In another study performed in a mouse model of melanoma and biopsies, β-catenin stabilization was associated with increased activation of MAPK/ERK and PI3K/AKT-signaling in BRAF activated and PTEN inactivated highly metastatic melanomas [98]. Full activation of AKT was detected, however, only in a small subset of melanomas [98]. These results suggest that the crosstalk between MAPK/ERK, PI3K/AKT and WNT/β-catenin-signaling depends on the genetic context. When phenotypic effects of the WNT/β-catenin pathway were compared in wild-type and mutated PTEN melanomas, the invasiveness and bioenergetics of melanoma cells were found to be regulated by WNT3A/β-catenin in a PTEN-dependent manner [57]. A most recent report has demonstrated that there is a high variability in the activity of PI3K/AKT and WNT/β-catenin pathways in melanomas with BRAFV600E and wild-type PTEN [143]. Moreover, the activity of these pathways can be largely modified in the response to targeted therapeutics and after the development of drug resistance [140,143,144]. Nuclear localization of β-catenin can be increased by prostaglandins, which indicates the potential crosstalk between WNT-signaling and the cyclooxygenase 2 (COX-2)/prostaglandin pathway [145]. COX-2 is a target gene of β-catenin transcription, and in addition, β-catenin contributes to the stabilization of COX-2 mRNA [145]. Transcription factors responding to diverse signal transduction pathways play a key role in integrating multiple signals to give rise to the optimal phenotype confronting the cancer developmental stage and microenvironmental insults [85]. MITF, a melanocyte and melanoma specific transcription factor is under control of many signaling pathways and active β-catenin contributes to its transcription [85]. MAPK/ERK, PI3K/AKT and WNT/β-catenin-signaling pathways are also able to post-translationally modify MITF [115]. ERK-mediated phosphorylation of MITF results in increased binding of p300/CBP [146] as well as proteasomal degradation [147,148]. It is worth to note that WNT/β-catenin, MAPK/ERK and PI3K/AKT-signaling can converge to regulate nuclear transport of MITF [149]. Phosphorylation of MITF by ERK1/2 primes MITF for phosphorylation by GSK3β [149], a kinase that is inhibited by both PI3K/AKT and WNT/β-catenin-signaling. This dual phosphorylation is an example how multiple signaling pathways can converge to control the activity of a specific transcription factor. MITF activity is also dependent on the non-canonical WNT pathway as PKC can activate STAT3 leading to inhibition of MITF expression with concomitant downregulation of melanocytic differentiation antigens (Melan A (MART1), DCT and gp100), thus promoting the metastatic phenotype [87,101]. The WNT5A impact on p53 activity is another example of the influence of non-canonical WNT-signaling on the transcription factor-mediated phenotype of melanoma cells. As shown most recently, a high expression of WNT5A is observed in wild-type p53 melanomas representing 80% of this cancer, and more important, WNT5A can promote a slow-cycling phenotype of melanoma cells by increasing the half-life of wild-type p53 [150]. WNT5A expression can be enhanced by aging or treatment of melanoma with targeted therapy or irradiation [82,121,151]. Following different types of stress, a slow-cycling phenotype is induced, which in turn may promote melanoma survival, plasticity and heterogeneity. However, WNT5A can stabilize p53, p53-induced apoptosis is inhibited, and melanoma cells are arrested. As p53 has been shown to inhibit WNT-signaling [152–154], a feedback loop WNT-p53-WNT that is possible in wild type p53 melanomas has been suggested [150]. A possible explanation is that PKC activated by WNT5A phosphorylates the inhibitor of apoptosis stimulating protein (iASPP) leading to its nuclear localization, which prevents p53-induced apoptosis [133,155]. As SIAH2 is a target of p53 [156,157], WNT5A-stabilized p53 may in turn increase SIAH2 expression and β-catenin degradation in melanoma [150]. Most recently, it has been shown that β-catenin can be a direct partner of SOX10 via protein–protein interactions, which reduces SOX10 protein level in melanoma cells [158]. These results suggest that SOX10 considered as functionally crucial for melanoma survival can be suppressed by WNT/β-catenin-signaling. Of note, SOX10 level has not been reduced by main targeted therapeutics used in melanoma treatment [117,158], and in selected vemurafenib- or -resistant melanoma cell lines has been even increased [143]. Interestingly, Int. J. Mol. Sci. 2020, 21, 4852 14 of 31

SOX10 expression in hepatocellular carcinoma has been shown to correlate with enhanced level of β-catenin, and active β-catenin forming a stable SOX10/TCF4/β-catenin complex is necessary for the oncogenic effects of SOX10 [159]. These examples illustrate how direct and indirect interactions of WNT-signaling with transcription factors can influence the melanoma cell phenotype.

5. WNT-Signaling in Cancer Immunity Various studies indicate that WNT-signaling is associated with several aspects of immunity, and alterations in WNT-signaling can be connected with the deregulation of immune response against cancer, both innate and adaptive [56,160–167]. Tumors can be classified as having or lacking a T-cell-inflamed microenvironment [168] and immunotherapeutic interventions, including checkpoint inhibitors such as antiprogrammed death 1 (PD-1), programmed death-ligand 1 (PD-L1) and anti-cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) antibodies have shown efficacy in patients with a preexisting T cell-inflamed cancer microenvironment [169]. Therefore, understanding the mechanisms underlying T cell exclusion is of clinical relevance. Most recently performed analysis of tumor microenvironment has revealed that about one-third of solid tumors have poor T cell infiltration, and tumor-intrinsic WNT/β-catenin-signaling inversely correlates with a T cell-inflamed phenotype in 90% of tumor types [170]. Authors suggest that activation of WNT/β-catenin in tumor cells is one of the possible mechanisms of intrinsic resistance to immunotherapy. The link between the WNT/β-catenin pathway and immune exclusion has been first identified in melanoma [171,172]. profiling and exome sequencing of 266 individual melanoma metastases has revealed that about 48% of the non-inflamed melanomas exert active WNT/β-catenin pathway with elevated expression of genes under control of this signaling [171]. Mutations potentially leading to the WNT/β-catenin pathway activation, either gain-of-function mutations in CTNNB1 (8%) or loss of function mutations (11%) in genes encoding negative regulators of WNT/β-catenin-signaling have been detected in these metastases [171]. Genetically engineered mouse models expressing conditionally active β-catenin have been used to show that melanomas with enhanced activation of WNT/β-catenin lack T cell infiltrate, which is due to insufficient recruitment of CD103/CD8α dendritic cells (DCs) [172]. This escape of immunity developed after activation of WNT-signaling in melanoma cells is accompanied by reduced of CC-motif chemokine ligand 4 (CCL4), a chemokine that attracts the immune cells (Figure5). The CCL4 expression is downregulated by β-catenin via activating transcription factor 3 (ATF3) [172]. Interestingly, no therapeutic effect of anti-CTLA-4 and anti-PD-L1 was observed in mice with melanoma expressing active β-catenin unless DCs were injected to activate tumor antigen-specific T cells. As immune evasion mediated by WNT/β-catenin-signaling frequently operates already in primary melanomas, it has been suggested that β-catenin-based stratification of patients may improve immunotherapeutic outcomes [173]. As activation of tumor-intrinsic WNT/β-catenin-signaling is enhanced in non-T cell-inflamed tumors, the WNT-signaling inhibitors may be useful for restoring immune cell infiltration to support immunotherapy. Int. J. Mol. Sci. 2020, 21, 4852 15 of 31 Int. J. Mol. Sci. 2020, 21, x FOR PEER REVIEW 15 of 32

Figure 5. Mechanisms of immune exclusion in melanoma through WNT/β-catenin-signaling [172]. β-catenin Figure 5. Mechanisms of immune exclusion in melanoma through WNT/β-catenin-signaling [172]. induces expression of ATF3 and ATF3 represses transcription of CCL4. CD103+ dendritic cells and cytotoxic β-catenin induces expression of ATF3 and ATF3 represses transcription of CCL4. CD103+ dendritic + CD8cells T-cells and are cytotoxic not recruited CD8+ T-cells to melanoma, are not recruitedwhich leads to melanoma, to non-inflamed which tumor. leadsto When non-inflamed β-catenin- tumor.signaling is inactiveWhen β in-catenin-signaling melanoma cells, isATF3 inactive is not in expressed, melanoma which cells, ATF3 restoresis not CCL4 expressed, production which and restores secretion. CCL4 This + + stimulatesproduction recruitment and secretion. of immune This stimulates cells, including recruitment CD103 of immune dendritic cells, including cells that CD103 activate+ dendritic CD8 cellsT cells. + Recruitmentthat activate of tumorCD8+ T specific cells. Recruitment CD8 T cells of intumor the specifictumor microenvironment CD8+ T cells in the results tumor microenvironment in immune inflamed melanoma.results inATF3 immune— activating inflamed transcription melanoma. factor ATF3—activating 3; β- cat— β- catenin; transcription CCL4 factor— CC- 3;motifβ- cat— chemokineβ- catenin; ligand 4. CCL4—CC-motif chemokine ligand 4.

6.As WNT-Targeted activation of tumor Therapies-intrinsic WNT/β-catenin-signaling is enhanced in non-T cell-inflamed tumors, the WNT-signaling inhibitors may be useful for restoring immune cell infiltration to support The important role of WNT-signaling in the development and adult life as well as its implication immunotherapy. in a wide spectrum of diseases, including cancer has already attracted the attention of medical and biotech companies. Ongoing research is focused on the development of specific inhibitors 6. WNT-Targeted Therapies of WNT/β-catenin-signaling for cancer therapies, and only very few modulators of non-canonical signalingThe important are known. role of Based WNT on-signaling the recent in advancesthe development in cancer and biology, adult targetinglife as well of as WNT-signaling its implication in in a widecancer spectrum cells has of to diseases, be put into including perspective cancer of hasits e ff alreadyectiveness attracted either in the reversing attention resistance of medical to anticancer and biotech companies.drugs or Ongoingin inhibition research of tumor is focused evasion. on It isthe important development to underline of specific that inhibitors it is difficult of to WNT target/β-catenin only - signalingthe WNT for pathwaycancer therapies, without and interfering only very with few other modulators signaling of pathways. non-canonical Diverse signaling compounds are known. of natural Based on originthe recent or synthetic, advances peptides in cancer and biology, antibodies targeting that are of in WNT preclinical-signaling studies in arecancer shown cells in Tablehas to2. be Natural put into perspectivecompounds of its targeting effectiveness WNT-signaling either in reversing have been resistance discussed to extensively anticancer [drugs174,175 or], in and inhibition therefore of only tumor evasion.few of It them is important are included to underline in Table 2that. it is difficult to target only the WNT pathway without interfering with other signaling pathways. Diverse compounds of natural origin or synthetic, peptides and antibodies that are in preclinical studies are shown in Table 2. Natural compounds targeting WNT-signaling have been discussed extensively [174,175], and therefore only few of them are included in Table 2.

Int. J. Mol. Sci. 2020, 21, 4852 16 of 31

Table 2. WNT/β -catenin pathway inhibitors in melanoma (preclinical study).

Target Compound Research Model References Mode of Action WM266 human melanoma cell line; porcupine inhibitor (synergizes B16/F10 mouse melanoma cell line; C59 with CTLA4-targeting antibodies [176] patient derived Hu-175, Hu-422, Hu-424, in mouse melanoma models) Hu-451 human melanoma cell lines WNT-2Ab WNT2 antibody LOX, FEMX melanoma cell lines; [177] FJ9 DVL inhibitor LOX melanoma cell line [178] inhibitor G007-LK B16/F10 mouse melanoma cell line [179] (sensitizes melanoma to PD-1 immune checkpoint blockade) (i) GSK3-β B16F10 mouse melanoma cell line [180] fisetin inhibitor (ii) activator of GSK3-β, DKK1 and 451Lu human melanoma cells; athymic [181] WIF-1; FZD and LRP5/6 inhibitor (nu/nu) female nude mice blocking the translocation of Mel 928, Mel 1241 and Mel 1011 human lupeol [182] β-catenin to the nucleus melanoma cell lines DMBC11, 12, 17, 19, 21 patient-derived pentoxifylline β-catenin [183] melanoma cell lines SKMEL28, A375, BLM, SKMEL19 and PKF115-584 β-catenin [100] 451Lu human melanoma cell lines

Several agents influencing WNT-signaling have reached the clinical trials (Table3). Many of these inhibitors cause, however, severe side effects associated with impairment of tissue homeostasis and tissue regeneration, and off-target effects of WNT inhibitor are still an unresolved problem. Most of the clinical trials are conducted with patients with colorectal cancer (Table3), and so far, melanoma patients were included only in one clinical trial (NCT01351103).

Table 3. Overview of clinical trials (current and past clinical trials) evaluating activity of drugs targeting WNT pathway in melanoma, colorectal cancer and hepatocellular carcinoma (https://clinicaltrials.gov).

Target/Mode of Compound Company Type of Cancer Trial Identifier (phase/status) Ref. Action WNT974 NCT02278133 metastatic colorectal (with LGX818 & Array BioPharma porcupine inhibitor (Phase 1; Phase 2/ completed; [184,185] cancer ) updated: October 2017) LGK974 BRAF mutant NCT01351103 (monotherapy or Novartis porcupine inhibitor colorectal cancer & (Phase 1/ [184,185] combined with Pharmaceuticals melanoma recruiting; updated: June 2020) PDR001) NCT02521844 ETC-1922159 EDDC, A*STAR porcupine inhibitor colorectal cancer (Phase 1/active, not recruiting; [186] (with pembrolizumab) Research Entities updated: October 2019) colorectal NCT03507998 adenocarcinoma CGX1321 Curegenix, Inc. porcupine inhibitor (Phase 1/ [187] hepatocellular recruiting; updated: May 2019) carcinoma NCT02675946 CGX1321 Curegenix, Inc. porcupine inhibitor colorectal cancer (Phase1/ [187] (with pembrolizumab) recruiting updated: September 2019) NCT01302405 Prism Pharma interaction of PRI-724 colorectal cancer (Phase 1 terminated due to low enrollment, [188] Co., Ltd. β-catenin and CBP updated: August 2017) DKN-01 NCT03645980 (monotherapy or Johannes Gutenberg hepatocellular DKK1 inhibitor (Phase 1; Phase 2/recruiting; updated: none combined with University Mainz carcinoma August 2019) ) OMP-54F28 (ipafricept) NCT02069145 FZD8 decoy locally advanced or with sorafenib OncoMed (Phase 1/completed; updated: August 2017) receptor for metastatic Pharmaceuticals, Inc. [189] OMP-54F28 WNT ligands hepatocellular cancer NCT01608867 (ipafricept) solid tumors (Phase 1/completed; updated: July 2017) Int. J. Mol. Sci. 2020, 21, 4852 17 of 31

Table 3. Cont.

Target/Mode of Compound Company Type of Cancer Trial Identifier (phase/status) Ref. Action FZD receptors NCT01345201 OMP-18R5 OncoMed (1, 2, 5, 7, 8) solid tumors (Phase 1/completed; [190] () Pharmaceuticals inhibitors updated: September 2016) NCT02655952 & colorectal cancer NCT02020291 Foxy-5 WntResearch AB WNT5A mimetic (Phase 1/completed; updated: December 2018) [191,192] Metastatic colorectal NCT03883802 cancer (Phase 2/recruiting; updated: April 2019) OMP-131R10 OncoMed anti-R-spondin3 NCT02482441 colorectal cancer [193] (with FOLFIRI) Pharmaceuticals, Inc. antibody (Phase 1/completed; updated: August 2018) NCT02687009 Michael Morse, MD FZD1 colorectal cancer [194–200] (Phase 1/terminated, updated: February 2020) internalization and niclosamide BCL-9 inhibition, NCT02519582 Charite University, metastatic colorectal c-JUN upregulation (Phase 2/ [201] Berlin, Germany cancer recruiting; updated: September 2018) NCT01985763 genistein Sofya Pintova GSK3-β inhibitor colorectal cancer [202,203] (Phase 1; Phase 2/completed; updated: May 2019) University of β-catenin/ NCT00256334 resveratrol colorectal cancer [204] California, Irvine TCF interaction (Phase 1/completed updated: June 2014) curcumin NCT01948661 (dietary supplements: Ente Ospedaliero β-catenin/ (Phase not applicable, trial without FDA- colorectal adenoma [205] Mirtoselect®®®® & Ospedali Galliera TCF interaction defined phases Meriva®®®®) /active, not recruiting; updated: August 2018) EDDC, Experimental Drug Development Center.

As shown in Tables2 and3, WNT pathways can be a ffected by drugs at various stages. The first stage, WNT ligand activation and binding, is the most intensively investigated part of the WNT/β-catenin pathway. Porcupine, an enzyme with the acyl-transferase activity necessary for palmitoylation of WNT ligands, was one of the first targets [6,8,14]. The porcupine inhibitor WNT974 (also known as LGK974) is under clinical evaluation in several cancer types, including melanoma (NCT01351103) [184,185] (Table3). C59 is another porcupine inhibitor investigated in melanoma that diminished WNT/β-catenin-signaling. It synergizes with anti-CLA-4 antibody in the B16 melanoma model, suggesting a synergistic enhancement in antitumor immunity [176]. A monoclonal antibody (WNT-2Ab) against human WNT2 ligand has been developed to induce apoptosis in melanoma cells exerting WNT2 overexpression, and WNT-2Ab treatment downregulated β-catenin target genes, e.g., CCND1 and c- [177]. Phase 1 clinical trial for Foxy-5 (formylated 6-amino-acid peptide fragment), a WNT5A-mimicking peptide has already been completed in colorectal cancer and has shown a promising therapeutic value [191,192]. FZD receptors are potential targets of WNT directed therapies. The attenuation of WNT-signaling can be achieved by ubiquitylation-mediated degradation of FZD by anti-RSPO3 mAb (OMP-131R10). Phase 1 clinical trial employing OMP-131R10 has been completed with colorectal cancer patients [193]. OMP-18R5 (Vantictumab), a human monoclonal antibody has been used to inhibit ligand binding by targeting FZD receptors (FZD 1, 2, 5, 7, 8) [190]. The activity of OMP-54F28 (Ipafricept, completed phase I clinical trial) is based on the competition with FZD8 for ligand binding. OMP-54F28 is a recombinant fusion protein that consists of cysteine-rich domain of FZD8 and Fc domain of immunoglobulin and it functions as a trap for FZD8-binding of WNTs [189,206]. FZD can also be targeted by niclosamide, a plant derived compound that is capable of inducing FZD1 internalization and DVL down-regulation [194,207]. It downregulates B-cell lymphoma 9 (BCL9) that impairs the formation of active β-catenin/TCF/LEF triple-complex and upregulates c-JUN [200]. Niclosamide has already entered clinical trials in colorectal cancer. Poor systemic bioavailability of niclosamide led to the development of pro-drugs with better pharmacokinetic properties [208]. DKK1 inhibitors also belong to molecules targeting WNT-signaling, as DKK1, initially characterized as tumor suppressor may also function as tumor promoter [209]. DKN-01, a humanized monoclonal neutralizing antibody against DKK1 recognized as having potential therapeutic implications [210], has entered clinical trials. There are also WNT-signaling inhibitors that target FZD-DVL interaction e.g., FJ9 that disrupts the interaction between FZD7 and DVL. FJ9 has been found to downregulate canonical WNT-signaling and induce apoptosis in and melanoma cells [178]. Another group of Int. J. Mol. Sci. 2020, 21, 4852 18 of 31 inhibitors belongs to intracellular drugs targeting the destruction complex. Tankyrases are the that degrade AXIN1 and AXIN2 through poly ADP-ribosylation. A very recent report indicates that inhibition of tankyrase by G007-LK can be used to overcome WNT/β-catenin-mediated resistance to immune checkpoint inhibitors [179]. While previously used tankyrase inhibitors caused bone loss [211] and intestinal toxicity [212], no such signs were observed in mice treated with G007-LK [179]. Growing evidence indicates that dietary factors along with alteration in the gutactivator protein 1 microbiota can affect WNT-signaling. Genistein, a soy-derived isoflavone, for which clinical trial phase 1 and 2 has been completed in CRC, inactivates WNT-signaling by GSK3-β targeting [202,203,213]. Fisetin (3,7,30,40-tetrahydroxyflavone) is a dietary flavonoid that inhibits GSK3-β and activates β-catenin in melanoma cells [180]. Another plant-derived molecule known to modulate WNT-signaling in melanoma is a triterpene lupeol, (lup-20(29)-en-3β-ol). It has been found that lupeol prevented the translocation of β-catenin to the nucleus, therefore decreased nuclear β-catenin level and expression of β-cateninInt. J. Mol. Sci. target 2020, 21, genesx FOR PEER e.g., REVIEW coding region determinant-binding protein (CRD-BP19 of), 32MITF and CCND1 [182]. Pentoxifylline, a drug approved by the FDA for the treatment of peripheral arterial β-catenin target genes e.g., coding region determinant-binding protein (CRD-BP), MITF and CCND1 [182]. disease,Pentoxifylline, markedly a reduced drug approved the level by the of active FDA forβ -catenin the treatment in the of nucleus peripheral of arterial melanoma disease, cells markedly with high basalreduced expression the leve of lβ of-catenin active β [-183catenin]. in the nucleus of melanoma cells with high basal expression of β- cateninAs demonstrated [183]. above, there is a large group of natural or synthetic compounds, peptides and antibodiesAs that demonstrated are capable above, of a ff thereecting, is a either large groupinducing of natural or inhibiting, or synthetic the WNT-signalingcompounds, peptides pathways and at antibodies that are capable of affecting, either inducing or inhibiting, the WNT-signaling pathways at various stages. Compounds that have been investigated in melanoma are shown in Figure6. various stages. Compounds that have been investigated in melanoma are shown in Figure 6.

Figure 6. Compounds and antibodies affecting WNT/β-catenin-signaling pathway that were investigated in Figure 6. Compounds and antibodies affecting WNT/β-catenin-signaling pathway that were investigated melanoma (preclinical studies, except for LGK974, which is tested in the clinical trial NCT01351103). APC— in melanomaadenomatosis (preclinical polyposis studies,coli; AXIN except—axis forinhibition LGK974, protein; which BRG is-1 tested—brahma in the-related clinical gene trial-1; BCL NCT01351103).-9—B-cell APC—adenomatosisCLL/lymphoma 9 protein; polyposis CBP coli;—cAMP AXIN—axis response inhibitionelement-binding protein; protein; BRG-1—brahma-related CK1α— gene-1;α; BCL-9—B-cellCK1γ—casein CLL kinase/lymphoma 1 γ; DKK1 9 protein;—Dickkopf CBP—cAMP-1; DVL— responsedisheveled; element-binding ER—endoplasmic protein; reticulum; CK1 FZDα—casein— kinasefrizzled; 1 α; GSK3β CK1γ——caseinglycogen kinasesynthase 1kinaseγ; DKK1—Dickkopf-1; 3β; LRP5/6—lipoprotein DVL—disheveled; receptor related protein ER—endoplasmic 5/6; PYGO— reticulum;pygopus; FZD—frizzled; TKNS—tankyrase; GSK3 β-TrCPβ——glycogenbeta-transducin synthase repeat–containing kinase 3 βprotein;; LRP5 Ub/6—lipoprotein— ubiquitin. receptor related protein 5/6; PYGO—pygopus; TKNS—tankyrase; β-TrCP—beta-transducin repeat–containing While inhibition of WNT-signaling represents an immensely appealing strategy for the development ofprotein; anticancer Ub—ubiquitin. therapeutics, none of drugs targeting WNT-signaling is yet available in clinical practice [4,214].

7. Conclusions WNT-signaling is extremely complex and context-dependent in cancer, including melanoma. Deregulation of WNT-signaling contributes to cancer initiation, progression, modulation of immune microenvironment and resistance to treatment. While WNT-signaling alterations start with APC mutations in about 70% of colorectal cancer patients, mutations are not the major cause of these deregulations in

Int. J. Mol. Sci. 2020, 21, 4852 19 of 31

While inhibition of WNT-signaling represents an immensely appealing strategy for the development of anticancer therapeutics, none of drugs targeting WNT-signaling is yet available in clinical practice [4,214].

7. Conclusions WNT-signaling is extremely complex and context-dependent in cancer, including melanoma. Deregulation of WNT-signaling contributes to cancer initiation, progression, modulation of immune microenvironment and resistance to treatment. While WNT-signaling alterations start with APC mutations in about 70% of colorectal cancer patients, mutations are not the major cause of these deregulations in melanoma. Moreover, transcriptionally active β-catenin is associated with less invasive disease and more favorable prognosis for melanoma patients, in contrast to other cancers, in which nuclear β-catenin is a driving force of both initiation and progression. It is thought that β-catenin-suppressed invasion occurs through a cell-type specific mechanism involving transcription factor MITF, one of the β-catenin target genes. On the other hand, low efficacy of immunotherapy is observed in melanomas with elevated level of β-catenin. The identification of therapeutic targets is further complicated by the crosstalk between WNT-signaling pathways and other signaling pathways crucial for melanoma development such as MAPK/ERK and PI3K/AKT, as well as the plasticity of melanoma cells in response to microenvironmental insults. Therefore, finding a therapeutic window for effective modulation of the WNT pathway in melanoma is a challenging task. A low number of clinical trials investigating WNT/β-catenin modulators in melanoma patients is the consequence of the controversial role of WNT-signaling in melanoma.

Author Contributions: A.G.-M. original draft preparation; M.C. supervised the conception of the work, review the literature and revised it critically. All authors have read and agreed to the published version of the manuscript. Funding: This work was financially supported by Medical University of Lodz (Grant No. 502-03/1-156-01/502-14-367 and 503/1-156-01/503-11-001). Acknowledgments: We would like to thank Markus Duechler and Marta Osrodek for helpful discussion. Conflicts of Interest: The authors declare no conflicts of interest.

Abbreviations

AP-1 activator protein 1 APC adenomatosis polyposis coli APE1 apurinic/apyrimidinic endonuclease 1 ARF6 adenosine diphosphate (ADP)-ribosylation factor 6 ARM armadillo ATF3 activating transcription factor 3 ATG5 autophagy-related gene 5 AXIN axis inhibition protein B9 L B-cell lymphoma 9-like BCL-9 B-cell CLL/lymphoma 9 protein β-TrCP beta-transducin repeat–containing protein BRG-1 brahma-related gene-1 CaMKII Ca2+/calmodulin dependent kinase II CaN calcineurin CAPN calpain CBP (CREB)-binding protein CCL4 CC-motif chemokine ligand 4 CDK2 cyclin dependent kinase 2 cer1 Cerberus protein CK1 γ casein kinase 1 γ CK1α casein kinase 1α CK1ε casein kinase 1ε CRC colorectal cancer Int. J. Mol. Sci. 2020, 21, 4852 20 of 31

CSCs cancer stem cells CTD C-terminal domain CTLA-4 anti-cytotoxic T lymphocyte-associated antigen 4 Daam-1 disheveled associated activator of morphogenesis 1 DAG diacylglycerol DCs dendritic cells DCT dopachrome tautomerase DKK Dickkopf DVL disheveled ER endoplasmic reticulum EVI/WLS Evenness interrupted/Wntless FGF fibroblast growth factor FOXOs forkhead box class O family member proteins FRA1 FOS-related antigen 1 FZD frizzled GSK3β glycogen synthase kinase 3β H3K9me methylation of histone H3 on lysine 9 HCC hepatocellular carcinoma iASPP inhibitor of apoptosis-stimulating protein of p53 ICAT inhibitor of β-catenin and TCF4 IL-β interleukin-1β IP3 inositol (1,4,5)-triphosphates IQGAP1 IQ IQ motif containing GTPase activating protein 1(IQGAP1) JNK c-Jun N-terminal kinases KRM Kremen LGR leucine-rich repeat-containing G-protein coupled receptor LGR5/RSPO G-protein coupled receptor 5/a roof plate-specific spondin LPPs lipoprotein particles LRP5/6 lipoprotein receptor related protein 5/6 MAK metastasis associated kinase MART1 melanoma antigen recognized by T cells 1 MITF microphthalmia-associated transcription factor MLC myosin light chain MMP matrix metalloproteinase NFAT nuclear factor of activated T-cells NLK Nemo-like kinase NPC nuclear pore complex NTD N-terminal domain NUPs nucleoporins OCT4 octamer-binding transcription factor 4 PAR1 protease-activated receptor 1 PCP planar cell polarity-signaling pathway PD-L1 programmed death-ligand 1 PIP2 phosphatidylinositol (4,5)-biphosphates PKC protein kinase C PLC phospholipase C PP2A 2A PPP(S/T)P Pro-Pro-Pro(SerTrp)Pro PTK7 protein tyrosine kinase 7 PYGO pygopus RNF43 ring finger protein 43 ROCK Rho-associated kinase ROR RAR-related orphan receptor RSPO R-spondin, roof plate-specific spondin Int. J. Mol. Sci. 2020, 21, 4852 21 of 31

RTK receptor tyrosine kinase RYK receptor like tyrosine kinase sFRPs secreted frizzled-related proteins SHF Src homology 2 domain containing F SIAH2 seven in absentia homolog 2 SLN sentinel lymph node SMAD mothers against decapentaplegic homolog SRY sex-determining region Y STAT3 Signal Transducer Moreover, Activator Of Transcription 3. SWI/SNF SWItch/Sucrose non-fermentable chromatin-remodeling complex TAK1 TGFβ-activated kinase 1 TCF/LEF T cell factor/lymphoid enhancer-binding factor 1 TGF-β transforming growth factor-β v-ATPase vacuolar H+-Adenosine Triphosphatase. VEGF vascular endothelial growth factor WIF1 WNT inhibitory factor 1 WISE WNT modulator in surface ectoderm WISP1 WNT1-inducible-signaling pathway protein 1 ZNRF3 zinc and ring finger protein 3

References

1. Nüsslein-Volhard, C.; Wieschaus, E. Mutations affecting segment number and polarity in Drosophila. Nature 1980, 287, 795–801. [CrossRef] 2. Nusse, R.; Varmus, H.E. Many tumors induced by the mouse mammary tumor virus contain a provirus integrated in the same region of the host genome. Cell 1982, 31, 99–109. [CrossRef] 3. Nusse, R.; Van Ooyen, A.; Cox, D.; Fung, Y.K.; Varmus, H. Mode of proviral activation of a putative mammary oncogene (int-1) on mouse chromosome 15. Nature 1984, 307, 131–136. [CrossRef] 4. Nusse, R.; Clevers, H. Wnt/β-Catenin Signaling, Disease, and Emerging Therapeutic Modalities. Cell 2017, 169, 985–999. [CrossRef] 5. Clevers, H. Wnt/beta-catenin signaling in development and disease. Cell 2006, 127, 469–480. [CrossRef] [PubMed] 6. Clevers, H.; Nusse, R. Wnt/β-catenin signaling and disease. Cell 2012, 149, 1192–1205. [CrossRef][PubMed] 7. Kahn, M. Can we safely target the WNT pathway? Nat. Rev. Drug Discov. 2014, 13, 513–532. [CrossRef] [PubMed] 8. Zhan, T.; Rindtorff, N.; Boutros, M. Wnt signaling in cancer. Oncogene 2017, 36, 1461–1473. [CrossRef] [PubMed] 9. Kumar, A.; Chalamalasetty, R.B.; Kennedy, M.W.; Thomas, S.; Inala, S.N.; Garriock, R.J.; Yamaguchi, T.P. Zfp703 Is a Wnt/β-Catenin Feedback Suppressor Targeting the β-Catenin/Tcf1 Complex. Mol. Cell. Biol. 2016, 36, 1793–1802. [CrossRef] 10. Grumolato, L.; Liu, G.; Mong, P.; Mudbhary, R.; Biswas, R.; Arroyave, R.; Vijayakumar, S.; Economides, A.N.; Aaronson, S.A. Canonical and noncanonical Wnts use a common mechanism to activate completely unrelated coreceptors. Genes Dev. 2010, 24, 2517–2530. [CrossRef] 11. Van Amerongen, R.; Mikels, A.; Nusse, R. Alternative wnt signaling is initiated by distinct receptors. Sci. Signal. 2008, 1, re9. [CrossRef][PubMed] 12. Amin, N.; Vincan, E. The Wnt signaling pathways and cell adhesion. Front. Biosci. 2012, 17, 784–804. [CrossRef][PubMed] 13. Pai, S.G.; Carneiro, B.A.; Mota, J.M.; Costa, R.; Leite, C.A.; Barroso-Sousa, R.; Kaplan, J.B.; Chae, Y.K.; Giles, F.J. Wnt/beta-catenin pathway: Modulating anticancer immune response. J. Hematol. Oncol. 2017, 10, 101. [CrossRef][PubMed] 14. Komiya, Y.; Habas, R. Wnt signal transduction pathways. Organogenesis 2008, 4, 68–75. [CrossRef][PubMed] 15. Buechling, T.; Boutros, M. Wnt signaling signaling at and above the receptor level. Curr. Top. Dev. Biol. 2011, 97, 21–53. Int. J. Mol. Sci. 2020, 21, 4852 22 of 31

16. O’Connell, M.P.; Weeraratna, A.T. Hear the Wnt Ror: How melanoma cells adjust to changes in Wnt. Pigment Cell Melanoma Res. 2009, 22, 724–739. [CrossRef] 17. Ackers, I.; Malgor, R. Interrelationship of canonical and non-canonical Wnt signalling pathways in chronic metabolic diseases. Diab. Vasc. Dis. Res. 2018, 15, 3–13. [CrossRef] 18. Mirabelli, C.K.; Nusse, R.; Tuveson, D.A.; Williams, B.O. Perspectives on the role of Wnt biology in cancer. Sci. Signal. 2019, 12, eaay4494. [CrossRef] 19. Corbett, L.; Mann, J.; Mann, D.A. Non-Canonical Wnt Predominates in Activated Rat Hepatic Stellate Cells, Influencing HSC Survival and Paracrine Stimulation of Kupffer Cells. PLoS ONE 2015, 10, e0142794. [CrossRef] 20. Anagnostou, S.H.; Shepherd, P.R. induces an autocrine activation of the Wnt/beta-catenin pathway in macrophage cell lines. Biochem. J. 2008, 416, 211–218. [CrossRef] 21. Tarapore, R.S.; Siddiqui, I.A.; Mukhtar, H. Modulation of Wnt/β-catenin signaling pathway by bioactive food components. 2012, 33, 483–491. [CrossRef][PubMed] 22. Van Dyke, T.; Merlino, G. β-catenin in metastatic melanoma—The smoking gun reloaded. Pigment Cell Melanoma Res. 2012, 25, 125–126. [CrossRef] 23. Jung, Y.S.; Park, J.I. Wnt signaling in cancer: Therapeutic targeting of Wnt signaling beyond β-catenin and the destruction complex. Exp. Mol. Med. 2020, 52, 183–191. [CrossRef][PubMed] 24. Liu, C.; Li, Y.; Semenov, M.; Han, C.; Baeg, G.H.; Tan, Y.; Zhang, Z.; Lin, X.; He, X. Control of beta-catenin phosphorylation/degradation by a dual-kinase mechanism. Cell 2002, 108, 837–847. [CrossRef] 25. Lee, E.; Salic, A.; Krüger, R.; Heinrich, R.; Kirschner, M.W. The roles of APC and Axin derived from experimental and theoretical analysis of the Wnt pathway. PLoS Biol. 2003, 1, E10. [CrossRef][PubMed] 26. Kim, M.J.; Chia, I.V.; Costantini, F. SUMOylation target sites at the C terminus protect Axin from ubiquitination and confer protein stability. FASEB J. 2008, 22, 3785–3794. [CrossRef][PubMed] 27. Mao, J.; Wang, J.; Liu, B.; Pan, W.; Farr, G.H., 3rd; Flynn, C.; Yuan, H.; Takada, S.; Kimelman, D.; Li, L.; et al. Low-density lipoprotein receptor-related protein-5 binds to Axin and regulates the canonical . Mol. Cell 2001, 7, 801–809. [CrossRef] 28. Huang, S.M.; Mishina, Y.M.; Liu, S.; Cheung, A.; Stegmeier, F.; Michaud, G.A.; Charlat, O.; Wiellette, E.; Zhang, Y.; Wiessner, S.; et al. Tankyrase inhibition stabilizes axin and antagonizes Wnt signalling. Nature 2009, 461, 614–620. [CrossRef] 29. Kim, S.E.; Huang, H.; Zhao, M.; Zhang, X.; Zhang, A.; Semonov,M.V.; MacDonald, B.T.; Zhang, X.; Garcia Abreu, J.; Peng, L.; et al. Wnt stabilization of β-catenin reveals principles for receptor-scaffold assemblies. Science 2013, 340, 867–870. [CrossRef] 30. Willert, K.; Shibamoto, S.; Nusse, R. Wnt-induced dephosphorylation of axin releases beta-catenin from the axin complex. Genes Dev. 1999, 13, 1768–1773. [CrossRef] 31. Ikeda, S.; Kishida, S.; Yamamoto, H.; Murai, H.; Koyama, S.; Kikuchi, A. Axin, a negative regulator of the Wnt signaling pathway, forms a complex with GSK-3beta and beta-catenin and promotes GSK-3beta-dependent phosphorylation of beta-catenin. EMBO J. 1998, 17, 1371–1384. [CrossRef][PubMed] 32. Han, J.I.; Na, K.J. Wnt/β-Catenin signaling pathway in canine skin melanoma and a possibility as a cancer model for human skin melanoma. In Melanoma in the Clinic Diagnosis, Management and Complications of Malignancy; Murph, M., Ed.; IntechOpen: London, UK, 2011. 33. Li, V.S.; Ng, S.S.; Boersema, P.J.; Low, T.Y.; Karthaus, W.R.; Gerlach, J.P.; Mohammed, S.; Heck, A.J.; Maurice, M.M.; Mahmoudi, T.; et al. Wnt signaling through inhibition of β-catenin degradation in an intact Axin1 complex. Cell 2012, 149, 1245–1256. [CrossRef][PubMed] 34. Uzdensky, A.B.; Demyanenko, S.V.; Bibov, M.Y. Signal transduction in human cutaneous melanoma and target drugs. Curr. Cancer Drug Targets 2013, 13, 843–866. [CrossRef] 35. Su, Y.; Fu, C.; Ishikawa, S.; Stella, A.; Kojima, M.; Shitoh, K.; Schreiber, E.M.; Day, B.W.; Liu, B. APC is essential for targeting phosphorylated beta-catenin to the SCFbeta-TrCP ubiquitin ligase. Mol. Cell 2008, 32, 652–661. [CrossRef] 36. Carmon, K.S.; Gong, X.; Lin, Q.; Thomas, A.; Liu, Q. R-spondins function as ligands of the orphan receptors LGR4 and LGR5 to regulate Wnt/beta-catenin signaling. Proc. Natl. Acad. Sci. USA 2011, 108, 11452–11457. [CrossRef][PubMed] Int. J. Mol. Sci. 2020, 21, 4852 23 of 31

37. De Lau, W.; Barker, N.; Low, T.Y.; Koo, B.K.; Li, V.S.; Teunissen, H.; Kujala, P.; Haegebarth, A.; Peters, P.J.; Van de Wetering, M.; et al. Lgr5 homologues associate with Wnt receptors and mediate R-spondin signalling. Nature 2011, 476, 293–297. [CrossRef][PubMed] 38. Glinka, A.; Dolde, C.; Kirsch, N.; Huang, Y.L.; Kazanskaya, O.; Ingelfinger, D.; Boutros, M.; Cruciat, C.M.; Niehrs, C. LGR4 and LGR5 are R-spondin receptors mediating Wnt/β-catenin and Wnt/PCP signalling. EMBO Rep. 2011, 12, 1055–1061. [CrossRef] 39. De Lau, W.; Peng, W.C.; Gros, P.; Clevers, H. The R-spondin/Lgr5/Rnf43 module: Regulator of Wnt signal strength. Genes Dev. 2014, 28, 305–316. [CrossRef] 40. Jamieson, C.; Sharma, M.; Henderson, B.R. Targeting the β-catenin nuclear transport pathway in cancer. Semin. Cancer Biol. 2014, 27, 20–29. [CrossRef] 41. Townsley, F.M.; Cliffe, A.; Bienz, M. Pygopus and Legless target Armadillo/beta-catenin to the nucleus to enable its transcriptional co-activator function. Nat. Cell Biol. 2004, 6, 626–633. [CrossRef] 42. Widlund, H.R.; Horstmann, M.A.; Price, E.R.; Cui, J.; Lessnick, S.L.; Wu, M.; He, X.; Fisher, D.E. Beta-catenin-induced melanoma growth requires the downstream target Microphthalmia-associated transcription factor. J. Cell Biol. 2002, 158, 1079–1087. [CrossRef] 43. Ring, A.; Kim, Y.M.; Kahn, M. Wnt/catenin signaling in physiology and disease. Stem Cell Rev. Rep. 2014, 10, 512–525. [CrossRef][PubMed] 44. Larue, L.; Delmas, V. The WNT/Beta-catenin pathway in melanoma. Front. Biosci. 2006, 11, 733–742. [CrossRef][PubMed] 45. Glinka, A.; Wu, W.; Delius, H.; Monaghan, A.P.; Blumenstock, C.; Niehrs, C. Dickkopf-1 is a member of a new family of secreted proteins and functions in head induction. Nature 1998, 391, 357–362. [CrossRef][PubMed] 46. Logan, C.Y.; Nusse, R. The Wnt signaling pathway in development and disease. Annu. Rev. Cell Dev. Biol. 2004, 20, 781–810. [CrossRef][PubMed] 47. Cici, D.; Corrado, A.; Rotondo, C.; Cantatore, F.P. Wnt Signaling and Biological Therapy in Rheumatoid Arthritis and Spondyloarthritis. Int. J. Mol. Sci. 2019, 20, E5552. [CrossRef] 48. Aktary,Z.; Bertrand, J.U.; Larue, L. The WNT-less wonder: WNT-independent β-catenin signaling. Pigment Cell Melanoma Res. 2016, 29, 524–540. [CrossRef] 49. Huber, A.H.; Weis, W.I. The structure of the beta-catenin/E-cadherin complex and the molecular basis of diverse ligand recognition by beta-catenin. Cell 2001, 105, 391–402. [CrossRef] 50. Valenta, T.; Hausmann, G.; Basler, K. The many faces and functions of β-catenin. EMBO J. 2012, 31, 2714–2736. [CrossRef] 51. Loh, C.Y.; Chai, J.Y.; Tang, T.F.; Wong, W.F.; Sethi, G.; Shanmugam, M.K.; Chong, P.P.; Looi, C.Y.The E-Cadherin and N-Cadherin Switch in Epithelial-to-Mesenchymal Transition: Signaling, Therapeutic Implications, and Challenges. Cells 2019, 8, E1118. [CrossRef] 52. Qian, D.; Jones, C.; Rzadzinska, A.; Mark, S.; Zhang, X.; Steel, K.P.; Dai, X.; Chen, P. Wnt5a functions in planar cell polarity regulation in mice. Dev. Biol. 2007, 306, 121–133. [CrossRef] 53. Kumawat, K.; Gosens, R. WNT-5A: Signaling and functions in health and disease. Cell. Mol. Life Sci. 2016, 73, 567–587. [CrossRef][PubMed] 54. Kulikova, K.; Kibardin, A.; Gnuchev, N.V.; Georgiev, G.P.; Larin, S. Wnt signaling pathway and its significance for melanoma development. CTM Mod. Technol. Med. 2012, 107–111. 55. Lang, C.M.R.; Chan, C.K.; Veltri, A.; Lien, W.H. Wnt signaling pathways in keratinocyte carcinomas. Cancers 2019, 11, E1216. [CrossRef][PubMed] 56. Galluzzi, L.; Spranger, S.; Fuchs, E.; López-Soto, A. WNT Signaling in Cancer Immunosurveillance. Trends Cell Biol. 2019, 29, 44–65. [CrossRef] 57. Brown, K.; Yang, P.; Salvador, D.; Kulikauskas, R.; Ruohola-Baker, H.; Robitaille, A.M.; Chien, A.J.; Moon, R.T.; Sherwood, V. WNT/β-catenin signaling regulates mitochondrial activity to alter the oncogenic potential of melanoma in a PTEN-dependent manner. Oncogene 2017, 36, 3119–3136. [CrossRef] 58. Kaplan, R.N.; Rafii, S.; Lyden, D. Preparing the “soil”: The premetastatic niche. Cancer Res. 2006, 66, 11089–11093. [CrossRef] 59. Psaila, B.; Lyden, D. The metastatic niche: Adapting the foreign soil. Nat. Rev. Cancer 2009, 9, 285–293. [CrossRef] 60. Sceneay, J.; Smyth, M.J.; Möller, A. The pre-metastatic niche: Finding common ground. Cancer Metastasis Rev. 2013, 32, 449–464. [CrossRef] Int. J. Mol. Sci. 2020, 21, 4852 24 of 31

61. Gajos-Michniewicz, A.; Czyz, M. Role of miRNAs in Melanoma Metastasis. Cancers 2019, 11, E326. [CrossRef] 62. Arozarena, I.; Bischof, H.; Gilby, D.; Belloni, B.; Dummer, R.; Wellbrock, C. In melanoma, beta-catenin is a suppressor of invasion. Oncogene 2011, 30, 4531–4543. [CrossRef][PubMed] 63. Schatoff, E.M.; Leach, B.I.; Dow, L.E. Wnt Signaling and Colorectal Cancer. Curr. Colorectal Cancer Rep. 2017, 13, 101–110. [CrossRef][PubMed] 64. White, B.D.; Chien, A.J.; Dawson, D.W. Dysregulation of Wnt/β-catenin signaling in gastrointestinal cancers. Gastroenterology 2012, 142, 219–232. [CrossRef][PubMed] 65. Kwong, L.N.; Dove, W.F. APC and its modifiers in colon cancer. Adv. Exp. Med. Biol. 2009, 656, 85–106. [PubMed] 66. Rowan, A.J.; Lamlum, H.; Ilyas, M.; Wheeler, J.; Straub, J.; Papadopoulou, A.; Bicknell, D.; Bodmer, W.F.; Tomlinson, I.P. APC mutations in sporadic colorectal tumors: A mutational “hotspot” and interdependence of the “two hits”. Proc. Natl. Acad. Sci. USA 2000, 97, 3352–3357. [CrossRef][PubMed] 67. Schell, M.J.; Yang, M.; Teer, J.K.; Lo, F.Y.; Madan, A.; Coppola, D.; Monteiro, A.N.; Nebozhyn, M.V.; Yue, B.; Loboda, A.; et al. A multigene mutation classification of 468 colorectal cancers reveals a prognostic role for APC. Nat. Commun. 2016, 7, 11743. [CrossRef] 68. Rubinfeld, B.; Robbins, P.; El-Gamil, M.; Albert, I.; Porfiri, E.; Polakis, P. Stabilization of beta-catenin by genetic defects in melanoma cell lines. Science 1997, 275, 1790–1792. [CrossRef] 69. Rimm, D.L.; Caca, K.; Hu, G.; Harrison, F.B.; Fearon, E.R. Frequent nuclear/cytoplasmic localization of beta-catenin without exon 3 mutations in malignant melanoma. Am. J. Pathol. 1999, 154, 325–329. [CrossRef] 70. Omholt, K.; Platz, A.; Ringborg, U.; Hansson, J. Cytoplasmic and nuclear accumulation of beta-catenin is rarely caused by CTNNB1 exon 3 mutations in cutaneous malignant melanoma. Int. J. Cancer 2001, 92, 839–842. [CrossRef] 71. Demunter, A.; Libbrecht, L.; Degreef, H.; De Wolf-Peeters, C.; Van den Oord, J.J. Loss of membranous expression of beta-catenin is associated with tumor progression in cutaneous melanoma and rarely caused by exon 3 mutations. Mod. Pathol. 2002, 15, 454–461. [CrossRef] 72. Pollock, P.M.; Hayward, N. Mutations in exon 3 of the beta-catenin gene are rare in melanoma cell lines. Melanoma Res. 2002, 12, 183–186. [CrossRef][PubMed] 73. Reifenberger, J.; Knobbe, C.B.; Wolter, M.; Blaschke, B.; Schulte, K.W.; Pietsch, T.; Ruzicka, T.; Reifenberger, G. Molecular genetic analysis of malignant melanomas for aberrations of the WNT signaling pathway genes CTNNB1, APC, ICAT and BTRC. Int. J. Cancer 2002, 100, 549–556. [CrossRef] 74. Worm, J.; Christensen, C.; Grønbaek, K.; Tulchinsky, E.; Guldberg, P. Genetic and epigenetic alterations of the APC gene in malignant melanoma. Oncogene 2004, 23, 5215–5226. [CrossRef] 75. Kuphal, S.; Lodermeyer, S.; Bataille, F.; Schuierer, M.; Hoang, B.H.; Bosserhoff, A.K. Expression of Dickkopf genes is strongly reduced in malignant melanoma. Oncogene 2006, 25, 5027–5036. [CrossRef] 76. Lin, Y.C.; You, L.; Xu, Z.; He, B.; Yang, C.T.; Chen, J.K.; Mikami, I.; Clément, G.; Shi, Y.; Kuchenbecker, K.; et al. Wnt inhibitory factor-1 gene transfer inhibits melanoma cell growth. Hum. Gene Ther. 2007, 18, 379–386. [CrossRef] 77. Huynh, K.T.; Takei, Y.; Kuo, C.; Scolyer, R.A.; Murali, R.; Chong, K.; Takeshima, L.; Sim, M.S.; Morton, D.L.; Turner, R.R.; et al. Aberrant hypermethylation in primary tumours and sentinel lymph node metastases in paediatric patients with cutaneous melanoma. Br. J. Dermatol. 2012, 166, 1319–1326. [CrossRef] 78. Luo, X.; Wei, B.; Chen, A.; Zhao, H.; Huang, K.; Chen, J. Methylation-mediated loss of SFRP2 enhances melanoma cell invasion via Wnt signaling. Am. J. Transl. Res. 2016, 8, 1502–1509. 79. Castiglia, D.; Bernardini, S.; Alvino, E.; Pagani, E.; De Luca, N.; Falcinelli, S.; Pacchiarotti, A.; Bonmassar, E.; Zambruno, G.; D’Atri, S. Concomitant activation of Wnt pathway and loss of mismatch repair function in human melanoma. Genes Cancer 2008, 47, 614–624. [CrossRef] 80. Aydin, I.T.; Melamed, R.D.; Adams, S.J.; Castillo-Martin, M.; Demir, A.; Bryk, D.; Brunner, G.; Cordon-Cardo, C.; Osman, I.; Rabadan, R.; et al. FBXW7 mutations in melanoma and a new therapeutic paradigm. J. Natl. Cancer Inst. 2014, 106, dju107. [CrossRef] 81. Balakrishnan, A.; Bleeker, F.E.; Lamba, S.; Rodolfo, M.; Daniotti, M.; Scarpa, A.; Van Tilborg, A.A.; Leenstra, S.; Zanon, C.; Bardelli, A. Novel somatic and germline mutations in cancer candidate genes in , melanoma, and pancreatic carcinoma. Cancer Res. 2007, 67, 3545–3550. [CrossRef] Int. J. Mol. Sci. 2020, 21, 4852 25 of 31

82. Kaur, A.; Webster, M.R.; Marchbank, K.; Behera, R.; Ndoye, A.; Kugel, C.H.; Dang, V.M.; Appleton, J.; O’Connell, M.P.; Cheng, P.; et al. sFRP2 in the aged microenvironment drives melanoma metastasis and therapy resistance. Nature 2016, 532, 250–254. [CrossRef] 83. Regad, T. Molecular and cellular pathogenesis of melanoma initiation and progression. Cell. Mol. Life Sci. 2013, 70, 4055–4065. [CrossRef] 84. Mort, R.L.; Jackson, I.J.; Patton, E.E. The melanocyte lineage in development and disease. Development 2015, 142, 620–632. [CrossRef] 85. Vance, K.W.; Goding, C.R. The transcription network regulating melanocyte development and melanoma. Pigment Cell Res. 2004, 17, 318–325. [CrossRef] 86. Schepsky, A.; Bruser, K.; Gunnarsson, G.J.; Goodall, J.; Hallsson, J.H.; Goding, C.R.; Steingrimsson, E.; Hecht, A. The microphthalmia-associated transcription factor Mitf interacts with beta-catenin to determine target gene expression. Mol. Cell. Biol. 2006, 26, 8914–8927. [CrossRef] 87. Webster, M.R.; Kugel, C.H.; Weeraratna, A.T. The Wnts of change: How Wnts regulate phenotype switching in melanoma. Biochim. Biophys. Acta 2015, 1856, 244–251. [CrossRef] 88. Lucero, O.M.; Dawson, D.W.; Moon, R.T.; Chien, A.J. A re-evaluation of the “oncogenic” nature of Wnt/beta-catenin signaling in melanoma and other cancers. Curr. Oncol. Rep. 2010, 12, 314–328. [CrossRef] 89. Silva, A.L.; Dawson, S.N.; Arends, M.J.; Guttula, K.; Hall, N.; Cameron, E.A.; Huang, T.H.; Brenton, J.D.; Tavaré, S.; Bienz, M.; et al. Boosting Wnt activity during colorectal cancer progression through selective hypermethylation of Wnt signaling antagonists. BMC Cancer 2014, 14, 891. [CrossRef] 90. Metcalfe, C.; Ibrahim, A.E.; Graeb, M.; De la Roche, M.; Schwarz-Romond, T.; Fiedler, M.; Winton, D.J.; Corfield, A.; Bienz, M. Dvl2 promotes intestinal length and neoplasia in the ApcMin mouse model for colorectal cancer. Cancer Res. 2010, 70, 6629–6638. [CrossRef] 91. Khalaf, A.M.; Fuentes, D.; Morshid, A.I.; Burke, M.R.; Kaseb, A.O.; Hassan, M.; Hazle, J.D.; Elsayes, K.M. Role of Wnt/β-catenin signaling in hepatocellular carcinoma, pathogenesis, and clinical significance. J. Hepatocell. Carcinoma 2018, 5, 61–73. [CrossRef] 92. Kim, E.; Lisby, A.; Ma, C.; Lo, N.; Ehmer, U.; Hayer, K.E.; Furth, E.E.; Viatour, P. Promotion of growth factor signaling as a critical function of β-catenin during HCC progression. Nat. Commun. 2019, 10, 1909. [CrossRef][PubMed] 93. Kovacs, D.; Migliano, E.; Muscardin, L.; Silipo, V.; Catricalà, C.; Picardo, M.; Bellei, B. The role of Wnt/β-catenin signaling pathway in melanoma epithelial-to-mesenchymal-like switching evidences from patients-derived cell lines. Oncotarget 2016, 7, 43295–43314. [CrossRef][PubMed] 94. Kageshita, T.; Hamby, C.V.; Ishihara, T.; Matsumoto, K.; Saida, T.; Ono, T. Loss of beta-catenin expression associated with disease progression in malignant melanoma. Br. J. Dermatol. 2001, 145, 210–216. [CrossRef] [PubMed] 95. Maelandsmo, G.M.; Holm, R.; Nesland, J.M.; Fodstad, Ø.; Flørenes, V.A. Reduced beta-catenin expression in the cytoplasm of advanced-stage superficial spreading malignant melanoma. Clin. Cancer Res. 2003, 9, 3383–3388. [PubMed] 96. Bachmann, I.M.; Straume, O.; Puntervoll, H.E.; Kalvenes, M.B.; Akslen, L.A. Importance of P-cadherin, beta-catenin, and Wnt5a/frizzled for progression of melanocytic tumors and prognosis in cutaneous melanoma. Clin. Cancer Res. 2005, 11, 8606–8614. [CrossRef] 97. Osborne, J.E. Loss of beta-catenin expression is associated with disease progression in malignant melanoma. Br. J. Dermatol. 2002, 146, 1104. [CrossRef] 98. Damsky, W.E.; Curley, D.P.; Santhanakrishnan, M.; Rosenbaum, L.E.; Platt, J.T.; Gould Rothberg, B.E.; Taketo, M.M.; Dankort, D.; Rimm, D.L.; McMahon, M.; et al. β-catenin signaling controls metastasis in Braf-activated Pten-deficient melanomas. Cancer Cell 2011, 20, 741–754. [CrossRef] 99. Gallagher, S.J.; Rambow, F.; Kumasaka, M.; Champeval, D.; Bellacosa, A.; Delmas, V.; Larue, L. Beta-catenin inhibits melanocyte migration but induces melanoma metastasis. Oncogene 2013, 32, 2230–2238. [CrossRef] 100. Sinnberg, T.; Levesque, M.P.; Krochmann, J.; Cheng, P.F.; Ikenberg, K.; Meraz-Torres, F.; Niessner, H.; Garbe, C.; Busch, C. Wnt-signaling enhances neural crest migration of melanoma cells and induces an invasive phenotype. Mol. Cancer 2018, 17, 59. [CrossRef] 101. Kaur, A.; Webster, M.R.; Weeraratna, A.T. In the Wnt-er of life: Wnt signalling in melanoma and ageing. Br. J. Cancer 2016, 115, 1273–1279. [CrossRef] Int. J. Mol. Sci. 2020, 21, 4852 26 of 31

102. Larue, L.; Beermann, F. Cutaneous melanoma in genetically modified . Pigment Cell Res. 2007, 20, 485–497. [CrossRef][PubMed] 103. Delmas, V.; Beermann, F.; Martinozzi, S.; Carreira, S.; Ackermann, J.; Kumasaka, M.; Denat, L.; Goodall, J.; Luciani, F.; Viros, A.; et al. Beta-catenin induces immortalization of melanocytes by suppressing p16INK4a expression and cooperates with N-Ras in melanoma development. Genes Dev. 2007, 21, 2923–2935. [CrossRef] [PubMed] 104. Larue, L.; Luciani, F.; Kumasaka, M.; Champeval, D.; Demirkan, N.; Bonaventure, J.; Delmas, V. Bypassing melanocyte senescence by beta-catenin: A novel way to promote melanoma. Pathol. Biol. 2009, 57, 543–547. [CrossRef][PubMed] 105. Collado, M.; Blasco, M.A.; Serrano, M. Cellular senescence in cancer and aging. Cell 2007, 130, 223–233. [CrossRef] 106. Goodall, J.; Martinozzi, S.; Dexter, T.J.; Champeval, D.; Carreira, S.; Larue, L.; Goding, C.R. Brn-2 expression controls melanoma proliferation and is directly regulated by beta-catenin. Mol. Cell. Biol. 2004, 24, 2915–2922. [CrossRef] 107. Chien, A.J.; Moore, E.C.; Lonsdorf, A.S.; Kulikauskas, R.M.; Rothberg, B.G.; Berger, A.J.; Major, M.B.; Hwang, S.T.; Rimm, D.L.; Moon, R.T. Activated Wnt/beta-catenin signaling in melanoma is associated with decreased proliferation in patient tumors and a murine melanoma model. Proc. Natl. Acad. Sci. USA 2009, 106, 1193–1198. [CrossRef] 108. Hoek, K.S.; Eichhoff, O.M.; Schlegel, N.C.; Döbbeling, U.; Kobert, N.; Schaerer, L.; Hemmi, S.; Dummer, R. In vivo switching of human melanoma cells between proliferative and invasive states. Cancer Res. 2008, 68, 650–656. [CrossRef] 109. Levy, C.; Khaled, M.; Fisher, D.E. MITF: Master regulator of melanocyte development and melanoma oncogene. Trends Mol. Med. 2006, 12, 406–414. [CrossRef] 110. Kawakami, A.; Fisher, D.E. The master role of microphthalmia-associated transcription factor in melanocyte and melanoma biology. Lab. Investig. 2017, 97, 649–656. [CrossRef] 111. Carreira, S.; Goodall, J.; Denat, L.; Rodriguez, M.; Nuciforo, P.; Hoek, K.S.; Testori, A.; Larue, L.; Goding, C.R. Mitf regulation of Dia1 controls melanoma proliferation and invasiveness. Genes Dev. 2006, 20, 3426–3439. [CrossRef] 112. Hoek, K.S.; Goding, C.R. Cancer stem cells versus phenotype-switching in melanoma. Pigment Cell Melanoma Res. 2010, 23, 746–759. [CrossRef] [PubMed] 113. Goding, C.R. Commentary. A picture of Mitf in melanoma immortality. Oncogene 2011, 30, 2304–2306. [CrossRef][PubMed] 114. Wellbrock, C.; Arozarena, I. Microphthalmia-associated transcription factor in melanoma development and MAP-kinase pathway targeted therapy. Pigment Cell Melanoma Res. 2015, 28, 390–406. [CrossRef] 115. Hartman, M.L.; Czyz, M. MITF in melanoma: Mechanisms behind its expression and activity. Cell. Mol. Life Sci. 2015, 72, 1249–1260. [CrossRef][PubMed] 116. Ahmed, F.; Haass, N.K. Microenvironment-Driven Dynamic Heterogeneity and Phenotypic Plasticity as a Mechanism of Melanoma Therapy Resistance. Front. Oncol. 2018, 8, 173. [CrossRef][PubMed] 117. Czyz, M.; Sztiller-Sikorska, M.; Gajos-Michniewicz, A.; Osrodek, M.; Hartman, M.L. Plasticity of Drug-Naïve and Vemurafenib- or Trametinib-Resistant Melanoma Cells in Execution of Differentiation/Pigmentation Program. J. Oncol. 2019, 2019, 1697913. [CrossRef] 118. Hartman, M.L.; Czyz, M. Pro-survival role of MITF in melanoma. J. Investig. Dermatol. 2015, 135, 352–358. [CrossRef] 119. Tsoi, J.; Robert, L.; Paraiso, K.; Galvan, C.; Sheu, K.M.; Lay, J.; Wong, D.J.L.; Atefi, M.; Shirazi, R.; Wang, X.; et al. Multi-stage Differentiation Defines Melanoma Subtypes with Differential Vulnerability to Drug-Induced Iron-Dependent Oxidative Stress. Cancer Cell 2018, 33, C890–C904. [CrossRef] 120. Arozarena, I.; Wellbrock, C. Targeting invasive properties of melanoma cells. FEBS J. 2017, 284, 2148–2162. [CrossRef] 121. Carreira, S.; Goodall, J.; Aksan, I.; La Rocca, S.A.; Galibert, M.D.; Denat, L.; Larue, L.; Goding, C.R. Mitf cooperates with Rb1 and activates p21Cip1 expression to regulate cell cycle progression. Nature 2005, 433, 764–769. [CrossRef] Int. J. Mol. Sci. 2020, 21, 4852 27 of 31

122. Webster, M.R.; Xu, M.; Kinzler, K.A.; Kaur, A.; Appleton, J.; O’Connell, M.P.; Marchbank, K.; Valiga, A.; Dang, V.M.; Perego, M.; et al. Wnt5A promotes an adaptive, senescent-like stress response, while continuing to drive invasion in melanoma cells. Pigment Cell Melanoma Res. 2015, 28, 184–195. [CrossRef] 123. Mithani, S.K.; Smith, I.M.; Califano, J.A. Use of integrative epigenetic and cytogenetic analyses to identify novel tumor-suppressor genes in malignant melanoma. Melanoma Res. 2011, 21, 298–307. [CrossRef] [PubMed] 124. Weeraratna, A.T.; Jiang, Y.; Hostetter, G.; Rosenblatt, K.; Duray, P.; Bittner, M.; Trent, J.M. Wnt5a signaling directly affects cell motility and invasion of metastatic melanoma. Cancer Cell 2002, 1, 279–288. [CrossRef] 125. Da Forno, P.D.; Pringle, J.H.; Hutchinson, P.; Osborn, J.; Huang, Q.; Potter, L.; Hancox, R.A.; Fletcher, A.; Saldanha, G.S. WNT5A expression increases during melanoma progression and correlates with outcome. Clin. Cancer Res. 2008, 14, 5825–5832. [CrossRef][PubMed] 126. Bittner, M.; Meltzer, P.; Chen, Y.; Jiang, Y.; Seftor, E.; Hendrix, M.; Radmacher, M.; Simon, R.; Yakhini, Z.; Ben-Dor, A.; et al. Molecular classification of cutaneous malignant melanoma by gene expression profiling. Nature 2000, 406, 536–540. [CrossRef] 127. O’Connell, M.P.; Fiori, J.L.; Xu, M.; Carter, A.D.; Frank, B.P.; Camilli, T.C.; French, A.D.; Dissanayake, S.K.; Indig, F.E.; Bernier, M.; et al. The orphan tyrosine kinase receptor, ROR2, mediates Wnt5A signaling in metastatic melanoma. Oncogene 2010, 29, 34–44. [CrossRef] 128. Sherwood, V.; Chaurasiya, S.K.; Ekström, E.J.; Guilmain, W.; Liu, Q.; Koeck, T.; Brown, K.; Hansson, K.; Agnarsdóttir, M.; Bergqvist, M.; et al. WNT5A-mediated β-catenin-independent signalling is a novel regulator of cancer cell metabolism. Carcinogenesis 2014, 35, 784–794. [CrossRef] 129. Ekström, E.J.; Bergenfelz, C.; Von Bülow, V.; Serifler, F.; Carlemalm, E.; Jönsson, G.; Andersson, T.; Leandersson, K. WNT5A induces release of exosomes containing pro-angiogenic and immunosuppressive factors from malignant melanoma cells. Mol. Cancer 2014, 13, 88. [CrossRef] 130. Ndoye, A.; Budina-Kolomets, A.; Kugel, C.H., 3rd; Webster, M.R.; Kaur, A.; Behera, R.; Rebecca, V.W.; Li, L.; Brafford, P.A.; Liu, Q.; et al. ATG5 Mediates a Positive Feedback Loop between Wnt Signaling and Autophagy in Melanoma. Cancer Res. 2017, 77, 5873–5885. [CrossRef] 131. McDonald, S.L.; Silver, A. The opposing roles of Wnt-5a in cancer. Br. J. Cancer 2009, 101, 209–214. [CrossRef] 132. Grossmann, A.H.; Yoo, J.H.; Clancy, J.; Sorensen, L.K.; Sedgwick, A.; Tong, Z.; Ostanin, K.; Rogers, A.; Grossmann, K.F.; Tripp, S.R.; et al. The small GTPase ARF6 stimulates β-catenin transcriptional activity during WNT5A-mediated melanoma invasion and metastasis. Sci. Signal. 2013, 6, ra14. [CrossRef][PubMed] 133. Dissanayake, S.K.; Wade, M.; Johnson, C.E.; O’Connell, M.P.; Leotlela, P.D.; French, A.D.; Shah, K.V.; Hewitt, K.J.; Rosenthal, D.T.; Indig, F.E.; et al. The Wnt5A/protein kinase C pathway mediates motility in melanoma cells via the inhibition of metastasis suppressors and initiation of an epithelial to mesenchymal transition. J. Biol. Chem. 2007, 282, 17259–17271. [CrossRef][PubMed] 134. Sato, A.; Yamamoto, H.; Sakane, H.; Koyama, H.; Kikuchi, A. Wnt5a regulates distinct signalling pathways by binding to Frizzled2. EMBO J. 2010, 29, 41–54. [CrossRef][PubMed] 135. Topol, L.; Jiang, X.; Choi, H.; Garrett-Beal, L.; Carolan, P.J.; Yang, Y. Wnt-5a inhibits the canonical Wnt pathway by promoting GSK-3-independent beta-catenin degradation. J. Cell Biol. 2003, 162, 899–908. [CrossRef] [PubMed] 136. O’Connell, M.P.; Marchbank, K.; Webster, M.R.; Valiga, A.A.; Kaur, A.; Vultur, A.; Li, L.; Herlyn, M.; Villanueva, J.; Liu, Q.; et al. Hypoxia induces phenotypic plasticity and therapy resistance in melanoma via the tyrosine kinase receptors ROR1 and ROR2. Cancer Discov. 2013, 3, 1378–1393. [CrossRef] 137. Webster, M.R.; Weeraratna, A.T. A Wnt-er migration: The confusing role of β-catenin in melanoma metastasis. Sci. Signal. 2013, 6, pe11. [CrossRef] 138. Li, C.; Nguyen, V.; Clark, K.N.; Zahed, T.; Sharkas, S.; Filipp, F.V.; Boiko, A.D. Down-regulation of FZD3 receptor suppresses growth and metastasis of human melanoma independently of canonical WNT signaling. Proc. Natl. Acad. Sci. USA 2019, 116, 4548–4557. [CrossRef] 139. Siemers, N.O.; Holloway,J.L.; Chang, H.; Chasalow, S.D.; Ross-MacDonald, P.B.; Voliva, C.F.; Szustakowski, J.D. Genome-wide association analysis identifies genetic correlates of immune infiltrates in solid tumors. PLoS ONE 2017, 12, e0179726. [CrossRef] 140. Biechele, T.L.; Kulikauskas, R.M.; Toroni, R.A.; Lucero, O.M.; Swift, R.D.; James, R.G.; Robin, N.C.; Dawson, D.W.; Moon, R.T.; Chien, A.J. Wnt/β-catenin signaling and AXIN1 regulate apoptosis triggered by inhibition of the mutant kinase BRAFV600E in human melanoma. Sci. Signal. 2012, 5, ra3. [CrossRef] Int. J. Mol. Sci. 2020, 21, 4852 28 of 31

141. Guardavaccaro, D.; Clevers, H. Wnt/β-catenin and MAPK signaling: Allies and enemies in different battlefields. Sci. Signal. 2012, 5, 15. [CrossRef] 142. Jeong, W.J.; Yoon, J.; Park, J.C.; Lee, S.H.; Lee, S.H.; Kaduwal, S.; Kim, H.; Yoon, J.B.; Choi, K.Y. Ras stabilization through aberrant activation of Wnt/β-catenin signaling promotes intestinal tumorigenesis. Sci. Signal. 2012, 5, ra30. [CrossRef] 143. Hartman, M.L.; Sztiller-Sikorska, M.; Gajos-Michniewicz, A.; Czyz, M. Dissecting Mechanisms of Melanoma Resistance to BRAF and MEK Inhibitors Revealed Genetic and Non-Genetic Patient- and Drug-Specific Alterations and Remarkable Phenotypic Plasticity. Cells 2020, 9, E142. [CrossRef][PubMed] 144. Chien, A.J.; Haydu, L.E.; Biechele, T.L.; Kulikauskas, R.M.; Rizos, H.; Kefford, R.F.; Scolyer, R.A.; Moon, R.T.; Long, G.V.Targeted BRAF inhibition impacts survival in melanoma patients with high levels of Wnt/β-catenin signaling. PLoS ONE 2014, 9, e94748. [CrossRef][PubMed] 145. Serini, S.; Fasano, E.; Piccioni, E.; Monego, G.; Cittadini, A.R.; Celleno, L.; Ranelletti, F.O.; Calviello, G. DHA induces apoptosis and differentiation in human melanoma cells in vitro: Involvement of HuR-mediated COX-2 mRNA stabilization and β-catenin nuclear translocation. Carcinogenesis 2012, 33, 164–173. [CrossRef] 146. Price, E.R.; Ding, H.F.; Badalian, T.; Bhattacharya, S.; Takemoto, C.; Yao, T.P.; Hemesath, T.J.; Fisher, D.E. Lineage-specific signaling in melanocytes. C- stimulation recruits p300/CBP to microphthalmia. J. Biol. Chem. 1998, 273, 17983–17986. [CrossRef] [PubMed] 147. Wu, M.; Hemesath, T.J.; Takemoto, C.M.; Horstmann, M.A.; Wells, A.G.; Price, E.R.; Fisher, D.Z.; Fisher, D.E. c-Kit triggers dual , which couple activation and degradation of the essential melanocyte factor Mi. Genes Dev. 2000, 14, 301–312. 148. Xu, W.; Gong, L.; Haddad, M.M.; Bischof, O.; Campisi, J.; Yeh, E.T.; Medrano, E.E. Regulation of microphthalmia- associated transcription factor MITF protein levels by association with the ubiquitin-conjugating enzyme hUBC9. Exp. Cell Res. 2000, 255, 135–143. [CrossRef] 149. Ngeow, K.C.; Friedrichsen, H.J.; Li, L.; Zeng, Z.; Andrews, S.; Volpon, L.; Brunsdon, H.; Berridge, G.; Picaud, S.; Fischer, R.; et al. BRAF/MAPK and GSK3 signaling converges to control MITF nuclear export. Proc. Natl. Acad. Sci. USA 2018, 115, E8668–E8677. [CrossRef] 150. Webster, M.R.; Fane, M.E.; Alicea, G.M.; Basu, S.; Kossenkov, A.V.; Marino, G.E.; Douglass, S.M.; Kaur, A.; Ecker, B.L.; Gnanapradeepan, K.; et al. Paradoxical Role for Wild-Type p53 in Driving Therapy Resistance in Melanoma. Mol. Cell 2020, 77, 633–644. 151. Behera, R.; Kaur, A.; Webster, M.R.; Kim, S.; Ndoye, A.; Kugel, C.H.; Alicea, G.M.; Wang, J.; Ghosh, K.; Cheng, P.; et al. Inhibition of Age-Related Therapy Resistance in Melanoma by Rosiglitazone-Mediated Induction of . Clin. Cancer Res. 2017, 23, 3181–3190. [CrossRef] 152. Levina, E.; Oren, M.; Ben-Ze’ev, A. Downregulation of beta-catenin by p53 involves changes in the rate of beta-catenin phosphorylation and Axin dynamics. Oncogene 2004, 23, 4444–4453. [CrossRef][PubMed] 153. Liu, J.; Stevens, J.; Rote, C.A.; Yost, H.J.; Hu, Y.; Neufeld, K.L.; White, R.L.; Matsunami, N. Siah-1 mediates a novel beta-catenin degradation pathway linking p53 to the adenomatous polyposis coli protein. Mol. Cell 2001, 7, 927–936. [CrossRef] 154. Sadot, E.; Geiger, B.; Oren, M.; Ben-Ze’ev, A. Down-regulation of beta-catenin by activated p53. Mol. Cell. Biol. 2001, 21, 6768–6781. [CrossRef] 155. Lu, M.; Breyssens, H.; Salter, V.; Zhong, S.; Hu, Y.; Baer, C.; Ratnayaka, I.; Sullivan, A.; Brown, N.R.; Endicott, J.; et al. Restoring p53 Function in Human Melanoma Cells by Inhibiting and Cyclin B1/CDK1-Phosphorylated Nuclear iASPP. Cancer Cell 2016, 30, 822–823. [CrossRef] 156. Chan, P.; Möller, A.; Liu, M.C.; Sceneay, J.E.; Wong, C.S.; Waddell, N.; Huang, K.T.; Dobrovic, A.; Millar, E.K.; O’Toole, S.A.; et al. The expression of the ubiquitin ligase SIAH2 (seven in absentia homolog 2) is mediated through gene copy number in and is associated with a basal-like phenotype and p53 expression. Breast Cancer Res. 2011, 13, R19. [CrossRef][PubMed] 157. Grishina, I.; Debus, K.; García-Limones, C.; Schneider, C.; Shresta, A.; García, C.; Calzado, M.A.; Schmitz, M.L. SIAH-mediated ubiquitination and degradation of acetyl-transferases regulate the p53 response and protein . Biochim. Biophys. Acta 2012, 1823, 2287–2296. [CrossRef][PubMed] 158. Uka, R.; Britschgi, C.; Krättli, A.; Matter, C.; Mihic, D.; Okoniewski, M.J.; Gualandi, M.; Stupp, R.; Cinelli, P.; Dummer, R.; et al. Temporal activation of WNT/β-catenin signaling is sufficient to inhibit SOX10 expression and block melanoma growth. Oncogene 2020, 39, 4132–4154. [CrossRef][PubMed] Int. J. Mol. Sci. 2020, 21, 4852 29 of 31

159. Zhou, D.; Bai, F.; Zhang, X.; Hu, M.; Zhao, G.; Zhao, Z.; Liu, R. SOX10 is a novel oncogene in hepatocellular carcinoma through Wnt/β-catenin/TCF4 cascade. Tumour Biol. 2014, 35, 9935–9940. [CrossRef][PubMed] 160. Hong, Y.; Manoharan, I.; Suryawanshi, A.; Majumdar, T.; Angus-Hill, M.L.; Koni, P.A.; Manicassamy, B.; Mellor, A.L.; Munn, D.H.; Manicassamy, S. β-catenin promotes regulatory T-cell responses in tumors by inducing vitamin A metabolism in dendritic cells. Cancer Res. 2015, 75, 656–665. [CrossRef][PubMed] 161. Alves, C.H.; Ober-Blöbaum, J.L.; Brouwers-Haspels, I.; Asmawidjaja, P.S.; Mus, A.M.; Razawy, W.; Molendijk, M.; Clausen, B.E.; Lubberts, E. Dendritic Cell-Specific Deletion of β-Catenin Results in Fewer Regulatory T-Cells without Exacerbating Autoimmune Collagen-Induced Arthritis. PLoS ONE 2015, 10, e0142972. [CrossRef] 162. Gattinoni, L.; Zhong, X.S.; Palmer, D.C.; Ji, Y.; Hinrichs, C.S.; Yu, Z.; Wrzesinski, C.; Boni, A.; Cassard, L.; Garvin, L.M.; et al. Wnt signaling arrests effector T cell differentiation and generates CD8+ memory stem cells. Nat. Med. 2009, 15, 808–813. [CrossRef][PubMed] 163. Swafford, D.; Manicassamy, S. Wnt signaling in dendritic cells: Its role in regulation of immunity and tolerance. Discov. Med. 2015, 19, 303–310. 164. Xu, Y.; Banerjee, D.; Huelsken, J.; Birchmeier, W.; Sen, J.M. Deletion of beta-catenin impairs T cell development. Nat. Immunol. 2003, 4, 1177–1182. [CrossRef] 165. Staal, F.J.; Luis, T.C.; Tiemessen, M.M. WNT signalling in the immune system: WNT is spreading its wings. Nat. Rev. Immunol. 2008, 8, 581–593. [CrossRef][PubMed] 166. Gattinoni, L.; Ji, Y.; Restifo, N.P. Wnt/beta-catenin signaling in T-cell immunity and cancer immunotherapy. Clin. Cancer Res. 2010, 16, 4695–4701. [CrossRef][PubMed] 167. Van Loosdregt, J.; Coffer, P.J. The Role of WNT Signaling in Mature T Cells: T Cell Factor Is Coming Home. J. Immunol. 2018, 201, 2193–2200. [CrossRef] 168. Trujillo, J.A.; Sweis, R.F.; Bao, R.; Luke, J.J. T Cell-Inflamed versus Non-T Cell-Inflamed Tumors: A Conceptual Framework for Cancer Immunotherapy Drug Development and Combination Therapy Selection. Cancer Immunol. Res. 2018, 6, 990–1000. [CrossRef] 169. Olson, D.J.; Luke, J.J. The T-cell-inflamed tumor microenvironment as a paradigm for immunotherapy drug development. Immunotherapy 2019, 11, 155–159. [CrossRef] 170. Luke, J.J.; Bao, R.; Sweis, R.F.; Spranger, S.; Gajewski, T.F. WNT/β-catenin Pathway Activation Correlates with Immune Exclusion across Human Cancers. Clin. Cancer Res. 2019, 25, 3074–3083. [CrossRef] 171. Spranger, S.; Bao, R.; Gajewski, T.F. Melanoma-intrinsic β-catenin signalling prevents anti-tumour immunity. Nature 2015, 523, 231–235. [CrossRef] 172. Spranger, S.; Gajewski, T.F. A new paradigm for tumor immune escape: β-catenin-driven immune exclusion. J. Immunother. Cancer 2015, 3, 43. [CrossRef] 173. Nsengimana, J.; Laye, J.; Filia, A.; O’Shea, S.; Muralidhar, S.; Po´zniak,J.; Droop, A.; Chan, M.; Walker, C.; Parkinson, L.; et al. β-Catenin-mediated immune evasion pathway frequently operates in primary cutaneous melanomas. J. Clin. Investig. 2018, 128, 2048–2063. [CrossRef][PubMed] 174. Gajos-Michniewicz, A.; Czyz, M. Modulation of WNT/β-catenin Pathway in Melanoma by Biologically Active Components Derived from Plants. Fitoterapia 2016, 109, 283–292. [CrossRef] 175. Sferrazza, G.; Corti, M.; Brusotti, G.; Pierimarchi, P.; Temporini, C.; Serafino, A.; Calleri, E. Nature-derived compounds modulating Wnt/β-catenin pathway: A preventive and therapeutic opportunity in neoplastic diseases. Acta Pharm. Sin. 2020, in press. [CrossRef] 176. Holtzhausen, A.; Zhao, F.; Evans, K.S.; Tsutsui, M.; Orabona, C.; Tyler, D.S.; Hanks, B.A. Melanoma-Derived Wnt5a Promotes Local Dendritic-Cell Expression of IDO and Immunotolerance: Opportunities for Pharmacologic Enhancement of Immunotherapy. Cancer Immunol. Res. 2015, 3, 1082–1095. [CrossRef] 177. You, L.; He, B.; Xu, Z.; Uematsu, K.; Mazieres, J.; Fujii, N.; Mikami, I.; Reguart, N.; McIntosh, J.K.; Kashani-Sabet, M.; et al. An anti-Wnt-2 monoclonal antibody induces apoptosis in malignant melanoma cells and inhibits tumor growth. Cancer Res. 2004, 64, 5385–5389. [CrossRef] 178. Fujii, N.; You, L.; Xu, Z.; Uematsu, K.; Shan, J.; He, B.; Mikami, I.; Edmondson, L.R.; Neale, G.; Zheng, J.; et al. An antagonist of protein-protein interaction suppresses beta-catenin-dependent tumor cell growth. Cancer Res. 2007, 67, 573–579. [CrossRef] 179. Waaler, J.; Mygland, L.; Tveita, A.; Strand, M.F.; Solberg, N.T.; Olsen, P.A.; Aizenshtadt, A.; Fauskanger, M.; Lund, K.; Brinch, S.A.; et al. Tankyrase inhibition sensitizes melanoma to PD-1 immune checkpoint blockade in syngeneic mouse models. Commun. Biol. 2020, 3, 196. [CrossRef][PubMed] Int. J. Mol. Sci. 2020, 21, 4852 30 of 31

180. Molagoda, I.M.N.; Karunarathne, W.A.H.M.; Park, S.R.; Choi, Y.H.; Park, E.K.; Jin, C.Y.; Yu, H.; Jo, W.S.; Lee, K.T.; Kim, G.Y. GSK-3β-Targeting Fisetin Promotes Melanogenesis in B16F10 Melanoma Cells and Zebrafish Larvae Through β-Catenin Activation. J. Mol. Sci. 2020, 21, 312. [CrossRef][PubMed] 181. Syed, D.N.; Afaq, F.; Maddodi, N.; Johnson, J.J.; Sarfaraz, S.; Ahmad, A.; Setaluri, V.; Mukhtar, H. Inhibition of human melanoma cell growth by the dietary flavonoid fisetin is associated with disruption of Wnt/β-catenin signaling and decreased Mitf levels. J. Investig. Dermatol. 2011, 131, 1291–1299. [CrossRef][PubMed] 182. Tarapore, R.S.; Siddiqui, I.A.; Saleem, M.; Adhami, V.M.; Spiegelman, V.S.; Mukhtar, H. Specific targeting of Wnt/β-catenin signaling in human melanoma cells by a dietary triterpene lupeol. Carcinogenesis 2010, 31, 1844–1853. [CrossRef] 183. Talar, B.; Gajos-Michniewicz, A.; Talar, M.; Chouaib, S.; Czyz, M. Pentoxifylline Inhibits WNT Signalling in β-Cateninhigh Patient-Derived Melanoma Cell Populations. PLoS ONE 2016, 11, e0158275. [CrossRef] [PubMed] 184. Bagheri, M.; Tabatabae Far, M.A.; Mirzaei, H.; Ghasemi, F. Evaluation of antitumor effects of aspirin and LGK974 drugs on cellular signaling pathways, cell cycle and apoptosis in colorectal cancer cell lines compared to oxaliplatin drug. Fundam. Clin. Pharmacol. 2020, 34, 51–64. [CrossRef] 185. Liu, J.; Pan, S.; Hsieh, M.H.; Ng, N.; Sun, F.; Wang, T.; Kasibhatla, S.; Schuller, A.G.; Li, A.G.; Cheng, D.; et al. Targeting Wnt-driven cancer through the inhibition of Porcupine by LGK974. Proc. Natl. Acad. Sci. USA 2013, 110, 20224–20229. [CrossRef][PubMed] 186. Madan, B.; Ke, Z.; Harmston, N.; Ho, S.Y.; Frois, A.O.; Alam, J.; Jeyaraj, D.A.; Pendharkar, V.; Ghosh, K.; Virshup, I.H.; et al. Wnt addiction of genetically defined cancers reversed by PORCN inhibition. Oncogene 2016, 35, 2197–2207. [CrossRef][PubMed] 187. Li, C.; Cao, J.; Zhang, N.; Tu, M.; Xu, F.; Wei, S.; Chen, X.; Xu, Y. Identification of RSPO2 Fusion Mutations and Target Therapy Using a Porcupine Inhibitor. Sci. Rep. 2018, 8, 14244. [CrossRef][PubMed] 188. El-Khoueiry, A.B.; Ning, Y.; Yang, D.; Cole, S.; Kahn, M.; Zoghbi, M.; Berg, J.; Fujimori, M.; Inada, T.; Kouji, H.; et al. A phase I first-in-human study of PRI-724 in patients (pts) with advanced solid tumors. J. Clin. Oncol. 2013, 31, 2501. [CrossRef] 189. Le, P.N.; McDermott, J.D.; Jimeno, A. Targeting the Wnt pathway in human cancers: Therapeutic targeting with a focus on OMP-54F28. Pharmacol. Ther. 2015, 146, 1–11. [CrossRef] 190. Gurney, A.; Axelrod, F.; Bond, C.J.; Cain, J.; Chartier, C.; Donigan, L.; Fischer, M.; Chaudhari, A.; Ji, M.; Kapoun, A.M.; et al. Wnt pathway inhibition via the targeting of Frizzled receptors results in decreased growth and tumorigenicity of human tumors. Proc. Natl. Acad. Sci. USA 2012, 109, 11717–11722. [CrossRef] 191. Osman, J.; Bellamkonda, K.; Liu, Q.; Andersson, T.; Sjölander, A. The WNT5A Agonist Foxy5 Reduces the Number of Colonic Cancer Stem Cells in a Xenograft Mouse Model of Human Colonic Cancer. Anticancer Res. 2019, 39, 1719–1728. [CrossRef] 192. Mehdawi, L.M.; Prasad, C.P.; Ehrnström, R.; Andersson, T.; Sjölander, A. Non-canonical WNT5A signaling up-regulates the expression of the tumor suppressor 15-PGDH and induces differentiation of colon cancer cells. Mol. Oncol. 2016, 10, 1415–1429. [CrossRef][PubMed] 193. Diamond, J.R.; Eckhardt, S.G.; Bendell, J.C.; Munster, P.; Morris, V.K.; Kopetz, S.; Cattaruzza, F.; Kapoun, A.M.; Dupont, J.; Faoro, L. A Phase 1a/b study of OMP-131R10, an anti-RSPO3 antibody, in advanced solid tumors and previously treated metastatic colorectal cancer (CRC). In Proceedings of the TAT 2016 Conference, Washington, DC, USA, 21–23 March 2016. 194. Osada, T.; Chen, M.; Yang, X.Y.; Spasojevic, I.; Vandeusen, J.B.; Hsu, D.; Clary, B.M.; Clay, T.M.; Chen, W.; Morse, M.A.; et al. Antihelminth compound niclosamide downregulates Wnt signaling and elicits antitumor responses in tumors with activating APC mutations. Cancer Res. 2011, 71, 4172–4182. [CrossRef] 195. Mook, R.A., Jr.; Wang, J.; Ren, X.R.; Piao, H.; Lyerly, H.K.; Chen, W. Identification of novel triazole inhibitors of Wnt/β-catenin signaling based on the Niclosamide chemotype. Bioorg. Med. Chem. Lett. 2019, 29, 317–321. [CrossRef][PubMed] 196. Leung, S.W.; Chou, C.J.; Huang, T.C.; Yang, P.M. An Integrated Bioinformatics Analysis Repurposes an Antihelminthic Drug Niclosamide for Treating HMGA2-Overexpressing Human Colorectal Cancer. Cancers 2019, 11, E1482. [CrossRef][PubMed] 197. Park, S.Y.; Kim, J.Y.; Choi, J.H.; Kim, J.H.; Lee, C.J.; Singh, P.; Sarkar, S.; Baek, J.H.; Nam, J.S. Inhibition of LEF1-Mediated DCLK1 by Niclosamide Attenuates Colorectal Cancer Stemness. Clin. Cancer Res. 2019, 25, 1415–1429. [CrossRef][PubMed] Int. J. Mol. Sci. 2020, 21, 4852 31 of 31

198. Bhattacharyya, J.; Ren, X.R.; Mook, R.A.; Wang, J.; Spasojevic, I.; Premont, R.T.; Li, X.; Chilkoti, A.; Chen, W. Niclosamide-conjugated polypeptide nanoparticles inhibit Wnt signaling and colon cancer growth. Nanoscale 2017, 9, 12709–12717. [CrossRef] 199. Ahn, S.Y.; Kim, N.H.; Lee, K.; Cha, Y.H.; Yang, J.H.; Cha, S.Y.; Cho, E.S.; Lee, Y.; Cha, J.S.; Cho, H.S.; et al. Niclosamide is a potential therapeutic for familial adenomatosis polyposis by disrupting Axin-GSK3 interaction. Oncotarget 2017, 8, 31842–31855. [CrossRef] 200. Monin, M.B.; Krause, P.; Stelling, R.; Bocuk, D.; Niebert, S.; Klemm, F.; Pukrop, T.; Koenig, S. The anthelmintic niclosamide inhibits colorectal cancer cell lines via modulation of the canonical and noncanonical Wnt signaling pathway. J. Surg. Res. 2016, 203, 193–205. [CrossRef] 201. Burock, S.; Daum, S.; Keilholz, U.; Neumann, K.; Walther, W.; Stein, U. Phase II trial to investigate the safety and efficacy of orally applied niclosamide in patients with metachronous or sychronous metastases of a colorectal cancer progressing after therapy: The NIKOLO trial. BMC Cancer 2018, 18, 297. [CrossRef] 202. Pintova, S.; Dharmupari, S.; Moshier, E.; Zubizarreta, N.; Ang, C.; Holcombe, R.F. Genistein combined with FOLFOX or FOLFOX- for the treatment of metastatic colorectal cancer: Phase I/II pilot study. Cancer Chemother. Pharmacol. 2019, 84, 591–598. [CrossRef] 203. Pintova, S.; Planutis, K.; Planutiene, M.; Holcombe, R.F. ME-143 Is Superior to Genistein in Suppression of WNT Signaling in Colon Cancer Cells. Anticancer Res. 2017, 37, 1647–1653. [PubMed] 204. Nguyen, A.V.; Martinez, M.; Stamos, M.J.; Moyer, M.P.; Planutis, K.; Hope, C.; Holcombe, R.F. Results of a phase I pilot clinical trial examining the effect of plant-derived resveratrol and grape powder on Wnt pathway target gene expression in colonic mucosa and colon cancer. Cancer Manag. Res. 2009, 1, 25–37. 205. Park, C.H.; Hahm, E.R.; Park, S.; Kim, H.K.; Yang, C.H. The inhibitory mechanism of curcumin and its derivative against beta-catenin/Tcf signaling. FEBS Lett. 2005, 579, 2965–2971. [CrossRef][PubMed] 206. Katoh, M.; Katoh, M. Molecular genetics and targeted therapy of WNT-related human diseases (Review). Int. J. Mol. Med. 2017, 40, 587–606. [CrossRef][PubMed] 207. Chen, M.; Wang, J.; Lu, J.; Bond, M.C.; Ren, X.R.; Lyerly, H.K.; Barak, L.S.; Chen, W. The anti-helminthic niclosamide inhibits Wnt/Frizzled1 signaling. Biochemistry 2009, 48, 10267–10274. [CrossRef][PubMed] 208. Mook, R.A.; Wang, J.; Ren, X.R.; Chen, M.; Spasojevic, I.; Barak, L.S.; Lyerly, H.K.; Chen, W. Structure-activity studies of Wnt/β-catenin inhibition in the Niclosamide chemotype: Identification of derivatives with improved drug exposure. Bioorg. Med. Chem. 2015, 23, 5829–5838. [CrossRef] 209. Kagey,M.H.; He, X. Rationale for targeting the Wnt signalling modulator Dickkopf-1 for oncology. Br. J. Pharmacol. 2017, 174, 4637–4650. [CrossRef] 210. Wall, J.A.; Klempner, S.J.; Arend, R.C. The anti-DKK1 antibody DKN-01 as an immunomodulatory combination partner for the treatment of cancer. Expert Opin. Investig. Drugs 2020, in press. [CrossRef] 211. Fujita, S.; Mukai, T.; Mito, T.; Kodama, S.; Nagasu, A.; Kittaka, M.; Sone, T.; Ueki, Y.; Morita, Y.Pharmacological inhibition of tankyrase induces bone loss in mice by increasing osteoclastogenesis. Bone 2018, 106, 156–166. [CrossRef] 212. Lau, T.; Chan, E.; Callow, M.; Waaler, J.; Boggs, J.; Blake, R.A.; Magnuson, S.; Sambrone, A.; Schutten, M.; Firestein, R.; et al. A Novel Tankyrase Small-Molecule Inhibitor Suppresses APC Mutation-Driven Colorectal Tumor Growth. Cancer Res. 2013, 73, 3132–3144. [CrossRef] 213. Cheng, X.; Xu, X.; Chen, D.; Zhao, F.; Wang, W. Therapeutic potential of targeting the Wnt/β-catenin signaling pathway in colorectal cancer. Biomed. Pharmacother. 2019, 110, 473–481. [CrossRef][PubMed] 214. Krishnamurthy, N.; Kurzrock, R. Targeting the Wnt/beta-catenin Pathway in Cancer: Update on Effectors and Inhibitors. Cancer Treat. Rev. 2018, 62, 50–60. [CrossRef][PubMed]

© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).